51
|
Ahnaou A, Drinkenburg WHIM. Sleep, neuronal hyperexcitability, inflammation and neurodegeneration: Does early chronic short sleep trigger and is it the key to overcoming Alzheimer's disease? Neurosci Biobehav Rev 2021; 129:157-179. [PMID: 34214513 DOI: 10.1016/j.neubiorev.2021.06.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 05/13/2021] [Accepted: 06/25/2021] [Indexed: 01/13/2023]
Abstract
Evidence links neuroinflammation to Alzheimer's disease (AD); however, its exact contribution to the onset and progression of the disease is poorly understood. Symptoms of AD can be seen as the tip of an iceberg, consisting of a neuropathological build-up in the brain of extracellular amyloid-β (Aβ) plaques and intraneuronal hyperphosphorylated aggregates of Tau (pTau), which are thought to stem from an imbalance between its production and clearance resulting in loss of synaptic health and dysfunctional cortical connectivity. The glymphatic drainage system, which is particularly active during sleep, plays a key role in the clearance of proteinopathies. Poor sleep can cause hyperexcitability and promote Aβ and tau pathology leading to systemic inflammation. The early neuronal hyperexcitability of γ-aminobutyric acid (GABA)-ergic inhibitory interneurons and impaired inhibitory control of cortical pyramidal neurons lie at the crossroads of excitatory/inhibitory imbalance and inflammation. We outline, with a prospective framework, a possible vicious spiral linking early chronic short sleep, neuronal hyperexcitability, inflammation and neurodegeneration. Understanding the early predictors of AD, through an integrative approach, may hold promise for reducing attrition in the late stages of neuroprotective drug development.
Collapse
Affiliation(s)
- A Ahnaou
- Dept. of Neuroscience Discovery, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, Beerse, B-2340, Belgium.
| | - W H I M Drinkenburg
- Dept. of Neuroscience Discovery, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, Beerse, B-2340, Belgium
| |
Collapse
|
52
|
Stoiljkovic M, Gutierrez KO, Kelley C, Horvath TL, Hajós M. TREM2 Deficiency Disrupts Network Oscillations Leading to Epileptic Activity and Aggravates Amyloid-β-Related Hippocampal Pathophysiology in Mice. J Alzheimers Dis 2021; 88:837-847. [PMID: 34120899 DOI: 10.3233/jad-210041] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Genetic mutations in triggering receptor expressed on myeloid cells-2 (TREM2) have been strongly associated with increased risk of developing Alzheimer's disease (AD) and other progressive dementias. In the brain, TREM2 protein is specifically expressed on microglia suggesting their active involvement in driving disease pathology. Using various transgenic AD models to interfere with microglial function through TREM2, several recent studies provided important data indicating a causal link between TREM2 and underlying amyloid-β (Aβ) and tau pathology. However, mechanisms by which TREM2 contributes to increased predisposition to clinical AD and influences its progression still remain largely unknown. OBJECTIVE Our aim was to elucidate the potential contribution of TREM2 on specific oscillatory dynamic changes associated with AD pathophysiology. METHODS Spontaneous and brainstem nucleus pontis oralis stimulation-induced hippocampal oscillation paradigm was used to investigate the impact of TREM2 haploinsufficiency TREM2(Het) or total deficiency TREM2(Hom) on hippocampal network function in wild-type and Aβ overproducing Tg2576 mice under urethane anesthesia. RESULTS Partial (TREM2(Het)) or total (TREM2(Hom)) deletion of TREM2 led to increased incidence of spontaneous epileptiform seizures in both wild-type and Tg2576 mice. Importantly, deficiency of TREM2 in Tg2576 mice significantly diminished power of theta oscillation in the hippocampus elicited by brainstem-stimulation compared to wild-type mice. However, it did not affect hippocampal theta-phase gamma-amplitude coupling significantly, since over a 60%reduction was found in coupling in Tg2576 mice regardless of TREM2 function. CONCLUSION Our findings indicate a role for TREM2-dependent microglial function in the hippocampal neuronal excitability in both wild type and Aβ overproducing mice, whereas deficiency in TREM2 function exacerbates disruptive effects of Aβ on hippocampal network oscillations.
Collapse
Affiliation(s)
- Milan Stoiljkovic
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Karel Otero Gutierrez
- Department of Neuroimmunology, Acute Neurology and Pain, Biogen Inc., Cambridge, MA, USA
| | - Craig Kelley
- Joint Biomedical Engineering Program, The State University of New York-Downstate and New York University-Tandon, Brooklyn, NY, USA
| | - Tamas L Horvath
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Mihály Hajós
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA.,Cognito Therapeutics, Cambridge, MA, USA
| |
Collapse
|
53
|
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
|
54
|
Dejakaisaya H, Kwan P, Jones NC. Astrocyte and glutamate involvement in the pathogenesis of epilepsy in Alzheimer's disease. Epilepsia 2021; 62:1485-1493. [PMID: 33971019 DOI: 10.1111/epi.16918] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) can increase the risk of epilepsy by up to 10-fold compared to healthy age-matched controls. However, the pathological mechanisms that underlie this increased risk are poorly understood. Because disruption in brain glutamate homeostasis has been implicated in both AD and epilepsy, this might play a mechanistic role in the pathogenesis of epilepsy in AD. Prior to the formation of amyloid beta (Aβ) plaques, the brain can undergo pathological changes as a result of increased production of amyloid precursor protein (APP) and Aβ oligomers. Impairments in the glutamate uptake ability of astrocytes due to astrogliosis are hypothesized to be an early event occurring before Aβ plaque formation. Astrogliosis may increase the susceptibility to epileptogenesis of the brain via accumulation of extracellular glutamate and resulting excitotoxicity. Here we hypothesize that Aβ oligomers and proinflammatory cytokines can cause astrogliosis and accumulation of extracellular glutamate, which then contribute to the pathogenesis of epilepsy in AD. In this review article, we consider the evidence supporting a potential role of dysfunction of the glutamate-glutamine cycle and the astrocyte in the pathogenesis of epilepsy in AD.
Collapse
Affiliation(s)
- Hattapark Dejakaisaya
- Department of Neuroscience, Central Clinical School, The Alfred Hospital, Monash University, Melbourne, Vic., Australia.,Faculty of Medicine and Public Health, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Patrick Kwan
- Department of Neuroscience, Central Clinical School, The Alfred Hospital, Monash University, Melbourne, Vic., Australia.,Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Vic., Australia
| | - Nigel C Jones
- Department of Neuroscience, Central Clinical School, The Alfred Hospital, Monash University, Melbourne, Vic., Australia.,Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Vic., Australia
| |
Collapse
|
55
|
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
|
56
|
Wang X, Loi SM, Foster E, Chen Z, Velakoulis D, Kwan P. Predictors of New-Onset Epilepsy in People With Younger-Onset Neurocognitive Disorders. Front Aging Neurosci 2021; 13:637260. [PMID: 33815091 PMCID: PMC8010684 DOI: 10.3389/fnagi.2021.637260] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/23/2021] [Indexed: 01/19/2023] Open
Abstract
Objective: People with neurocognitive disorders (NCDs) have an increased risk of epilepsy. However, most studies investigating the risk of seizures in people with NCDs are limited to those with Alzheimer's disease (AD) and vascular dementia (VD), and those who developed dementia after age 65 years. A knowledge gap exists regarding factors associated with development of epilepsy in people with younger-onset NCD, and those with non-AD and non-VD dementia subtypes. In this study, we aimed to identify the factors associated with the development of epilepsy in people with younger-onset NCDs of varied etiologies, the majority of whom had symptom onset prior to age 65 years. Participants and Methods: This was a retrospective study reviewing the medical records of consecutive people admitted with cognitive impairment to a tertiary neuropsychiatry unit between 1 January 2004 and 30 April 2019. People diagnosed with primary NCDs were included in the analysis. The prevalence and characteristics of epilepsy were described. The factors associated with developing epilepsy were identified in a binary logistic regression model. Results: A total of 427 people were included. One hundred fourteen had Alzheimer's disease, 104 frontotemporal dementia, 51 vascular dementia, 69 movement disorder-associated dementia, and 89 unspecified NCD. The median age on admission was 59 years (range 33-86) and 75.2% (n = 321/427) had young-onset NCD with onset before 65 years of age. 40/427 (9.4%) people had epilepsy, and epilepsy onset clustered between 2 years before and 6 years after the onset of cognitive decline in 80% (n = 32/40). The most frequent seizure type was focal to bilateral tonic-clonic seizure (35%, n = 14/40). Most of the people (94.7%, n = 36/38) achieved seizure freedom with one or two antiseizure medications. People with unspecified NCD (compared to frontotemporal dementia and movement disorder-associated dementia, age of onset of NCDs ≤50 years, and current smoking status were independently associated with higher risk of developing epilepsy. Conclusion: Epilepsy is common in people with younger-onset NCDs, and a high index of suspicion is warranted particularly for those with unspecified subtype and smoking status. Smoking reduction or cessation should be further investigated as a potentially modifiable factor for risk reduction.
Collapse
Affiliation(s)
- Xinshi Wang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Neuroscience, The Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Neurology, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia
| | - Samantha M Loi
- Neuropsychiatry, The Royal Melbourne Hospital and Melbourne Neuropsychiatry Center, The University of Melbourne and The Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Emma Foster
- Department of Neuroscience, The Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Neurology, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia
| | - Zhibin Chen
- Department of Neuroscience, The Central Clinical School, Monash University, Melbourne, VIC, Australia.,School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Dennis Velakoulis
- Neuropsychiatry, The Royal Melbourne Hospital and Melbourne Neuropsychiatry Center, The University of Melbourne and The Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Patrick Kwan
- Department of Neuroscience, The Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Neurology, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia.,School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
57
|
Alcantara-Gonzalez D, Chartampila E, Criscuolo C, Scharfman HE. Early changes in synaptic and intrinsic properties of dentate gyrus granule cells in a mouse model of Alzheimer's disease neuropathology and atypical effects of the cholinergic antagonist atropine. Neurobiol Dis 2021; 152:105274. [PMID: 33484828 DOI: 10.1016/j.nbd.2021.105274] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [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
|
58
|
Sleep disorders and late-onset epilepsy of unknown origin: Understanding new trajectories to brain amyloidopathy. Mech Ageing Dev 2021; 194:111434. [PMID: 33444630 DOI: 10.1016/j.mad.2021.111434] [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: 10/28/2020] [Revised: 12/31/2020] [Accepted: 01/07/2021] [Indexed: 12/16/2022]
Abstract
The intertwining between epilepsy, sleep disorders and beta amyloid pathology has been progressively highlighted, as early identification and stratification of patients at high risk of cognitive decline is the need of the hour. Modification of the sleep-wake activity, such as sleep impairment or excessive daytime sleepiness, can critically affect cerebral beta amyloid levels. Both mice models and human studies have demonstrated a substantial increase in the burden of beta amyloid pathology after sleep-deprivation, with potential negative effects partially restored by sleep recovery. The accumulation of beta amyloid has been shown to be an early event in the course of Alzheimer's disease dementia. Beta amyloid accumulation has been linked to epileptic seizures epileptic seizures, with beta amyloid being itself pro-epileptogenic in mice models already at oligomeric stage, well before plaque deposition. Further supporting a potential relationship between beta amyloid and epilepsy: i) seizures happen in 1 out of oofut 10 patients with Alzheimer's disease in the prodromal stage, ii) epileptic activity accelerates cognitive decline in Alzheimer's disease, iii) people with late-onset epilepsy present a critically high risk of developing dementia. In this Review we highlight the role of beta amyloid as a potential shared mechanisms between sleep disorders, late-onset epilepsy, and cognitive decline.
Collapse
|
59
|
Lam AD, Noebels J. Night Watch on the Titanic: Detecting Early Signs of Epileptogenesis in Alzheimer Disease. Epilepsy Curr 2020; 20:369-374. [PMID: 33081517 PMCID: PMC7818196 DOI: 10.1177/1535759720964775] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Aberrant cortical network excitability is an inextricable feature of Alzheimer disease (AD) that can negatively impact memory and accelerate cognitive decline. Surface electroencephalogram spikes and intracranial recordings of nocturnal silent seizures in human AD, coupled with the abnormal neural synchrony that precedes development of behavioral seizures in mouse AD models, build the case for epileptogenesis as an early therapeutic target for AD. Since most individuals with AD do not develop overt seizures, leveraging functional biomarkers of epilepsy risk to stratify a heterogeneous AD patient population for treatment is research priority for successful clinical trial design. Who will benefit from antiseizure interventions, which one, and when should it begin?
Collapse
Affiliation(s)
- Alice D. Lam
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Jeffrey Noebels
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
60
|
Botterill JJ, Lu YL, LaFrancois JJ, Bernstein HL, Alcantara-Gonzalez D, Jain S, Leary P, Scharfman HE. An Excitatory and Epileptogenic Effect of Dentate Gyrus Mossy Cells in a Mouse Model of Epilepsy. Cell Rep 2020; 29:2875-2889.e6. [PMID: 31775052 PMCID: PMC6905501 DOI: 10.1016/j.celrep.2019.10.100] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 09/25/2019] [Accepted: 10/24/2019] [Indexed: 12/20/2022] Open
Abstract
The sparse activity of hippocampal dentate gyrus (DG) granule cells (GCs) is thought to be critical for cognition and behavior, whereas excessive DG activity may contribute to disorders such as temporal lobe epilepsy (TLE). Glutamatergic mossy cells (MCs) of the DG are potentially critical to normal and pathological functions of the DG because they can regulate GC activity through innervation of GCs or indirectly through GABAergic neurons. Here, we test the hypothesis that MC excitation of GCs is normally weak, but under pathological conditions, MC excitation of GCs is dramatically strengthened. We show that selectively inhibiting MCs during severe seizures reduced manifestations of those seizures, hippocampal injury, and chronic epilepsy. In contrast, selectively activating MCs was pro-convulsant. Mechanistic in vitro studies using optogenetics further demonstrated the unanticipated ability of MC axons to excite GCs under pathological conditions. These results demonstrate an excitatory and epileptogenic effect of MCs in the DG.
Collapse
Affiliation(s)
- Justin J Botterill
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Yi-Ling Lu
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - John J LaFrancois
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Hannah L Bernstein
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA; Department of Neuroscience & Physiology, New York University Langone Health, New York, NY 10016, USA
| | - David Alcantara-Gonzalez
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Swati Jain
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Paige Leary
- 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 Neuroscience & Physiology, New York University Langone Health, New York, NY 10016, USA; Department of Psychiatry, New York University Langone Health, New York, NY 10016, USA.
| |
Collapse
|
61
|
Mander BA. Local Sleep and Alzheimer's Disease Pathophysiology. Front Neurosci 2020; 14:525970. [PMID: 33071726 PMCID: PMC7538792 DOI: 10.3389/fnins.2020.525970] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 09/01/2020] [Indexed: 12/11/2022] Open
Abstract
Even prior to the onset of the prodromal stages of Alzheimer's disease (AD), a constellation of sleep disturbances are apparent. A series of epidemiological studies indicate that multiple forms of these sleep disturbances are associated with increased risk for developing mild cognitive impairment (MCI) and AD, even triggering disease onset at an earlier age. Through the combination of causal manipulation studies in humans and rodents, as well as targeted examination of sleep disturbance with respect to AD biomarkers, mechanisms linking sleep disturbance to AD are beginning to emerge. In this review, we explore recent evidence linking local deficits in brain oscillatory function during sleep with local AD pathological burden and circuit-level dysfunction and degeneration. In short, three deficits in the local expression of sleep oscillations have been identified in relation to AD pathophysiology: (1) frequency-specific frontal deficits in slow wave expression during non-rapid eye movement (NREM) sleep, (2) deficits in parietal sleep spindle expression, and (3) deficits in the quality of electroencephalographic (EEG) desynchrony characteristic of REM sleep. These deficits are noteworthy since they differ from that seen in normal aging, indicating the potential presence of an abnormal aging process. How each of these are associated with β-amyloid (Aβ) and tau pathology, as well as neurodegeneration of circuits sensitive to AD pathophysiology, are examined in the present review, with a focus on the role of dysfunction within fronto-hippocampal and subcortical sleep-wake circuits. It is hypothesized that each of these local sleep deficits arise from distinct network-specific dysfunctions driven by regionally-specific accumulation of AD pathologies, as well as their associated neurodegeneration. Overall, the evolution of these local sleep deficits offer unique windows into the circuit-specific progression of distinct AD pathophysiological processes prior to AD onset, as well as their impact on brain function. This includes the potential erosion of sleep-dependent memory mechanisms, which may contribute to memory decline in AD. This review closes with a discussion of the remaining critical knowledge gaps and implications of this work for future mechanistic studies and studies implementing sleep-based treatment interventions.
Collapse
Affiliation(s)
- Bryce A. Mander
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, United States
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
62
|
Johnson ECB, Ho K, Yu GQ, Das M, Sanchez PE, Djukic B, Lopez I, Yu X, Gill M, Zhang W, Paz JT, Palop JJ, Mucke L. Behavioral and neural network abnormalities in human APP transgenic mice resemble those of App knock-in mice and are modulated by familial Alzheimer's disease mutations but not by inhibition of BACE1. Mol Neurodegener 2020; 15:53. [PMID: 32921309 PMCID: PMC7489007 DOI: 10.1186/s13024-020-00393-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most frequent and costly neurodegenerative disorder. Although diverse lines of evidence suggest that the amyloid precursor protein (APP) is involved in its causation, the precise mechanisms remain unknown and no treatments are available to prevent or halt the disease. A favorite hypothesis has been that APP contributes to AD pathogenesis through the cerebral accumulation of the amyloid-β peptide (Aβ), which is derived from APP through sequential proteolytic cleavage by BACE1 and γ-secretase. However, inhibitors of these enzymes have failed in clinical trials despite clear evidence for target engagement. METHODS To further elucidate the roles of APP and its metabolites in AD pathogenesis, we analyzed transgenic mice overexpressing wildtype human APP (hAPP) or hAPP carrying mutations that cause autosomal dominant familial AD (FAD), as well as App knock-in mice that do not overexpress hAPP but have two mouse App alleles with FAD mutations and a humanized Aβ sequence. RESULTS Although these lines of mice had marked differences in cortical and hippocampal levels of APP, APP C-terminal fragments, soluble Aβ, Aβ oligomers and age-dependent amyloid deposition, they all developed cognitive deficits as well as non-convulsive epileptiform activity, a type of network dysfunction that also occurs in a substantive proportion of humans with AD. Pharmacological inhibition of BACE1 effectively reduced levels of amyloidogenic APP C-terminal fragments (C99), soluble Aβ, Aβ oligomers, and amyloid deposits in transgenic mice expressing FAD-mutant hAPP, but did not improve their network dysfunction and behavioral abnormalities, even when initiated at early stages before amyloid deposits were detectable. CONCLUSIONS hAPP transgenic and App knock-in mice develop similar pathophysiological alterations. APP and its metabolites contribute to AD-related functional alterations through complex combinatorial mechanisms that may be difficult to block with BACE inhibitors and, possibly, also with other anti-Aβ treatments.
Collapse
Affiliation(s)
- Erik C. B. Johnson
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158 USA
| | - Kaitlyn Ho
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
| | - Gui-Qiu Yu
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
| | - Melanie Das
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
| | - Pascal E. Sanchez
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
| | - Biljana Djukic
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
| | - Isabel Lopez
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
| | - Xinxing Yu
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
| | - Michael Gill
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
| | - Weiping Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Institute of Endocrinology, Tianjin Medical University Metabolic Diseases Hospital, Tianjin, China
| | - Jeanne T. Paz
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158 USA
| | - Jorge J. Palop
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158 USA
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158 USA
| |
Collapse
|
63
|
Jin N, Ziyatdinova S, Gureviciene I, Tanila H. Response of spike-wave discharges in aged APP/PS1 Alzheimer model mice to antiepileptic, metabolic and cholinergic drugs. Sci Rep 2020; 10:11851. [PMID: 32678276 PMCID: PMC7366932 DOI: 10.1038/s41598-020-68845-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 07/02/2020] [Indexed: 01/19/2023] Open
Abstract
Epileptic nonconvulsive spike-wave discharges (SWDs) are commonly seen in amyloid plaque bearing transgenic mice but only rarely in their wild-type littermates. To shed light on their possible treatment options, we assessed the effect of drugs with variable and known mechanisms of action on the occurrence of SWDs in aged APPswe/PS1dE9 mice. The treatments included prototypic antiepileptic drugs (ethosuximide and levetiracetam), donepezil as the typical Alzheimer drug and atropine as an antagonistic effect, GABAB antagonist CGP-35348, and alternate energy substrates beta-hydroxybutyrate (BHB), pyruvate and lactate on the occurrence of SWDs in aged APPswe/PS1dE9 mice. All agents were administered by single intraperitoneal injections at doses earlier documented to be effective and response was assessed by recording 3 h of video-EEG. Atropine at 25 mg/kg significantly decreased SWD occurrence in all behavioral states, and also resulted in altered frequency composition of SWDs and general EEG slowing during sleep. Ethosuximide at 200 mg/kg and levetiracetam at 75 mg/kg effectively suppressed SWDs only during a period of mixed behavioral states, but levetiracetam also increased SWDs in sleep. BHB at 1 g/kg decreased SWDs in sleep, while both pyruvate and lactate at the same dose tended to increase SWD number and total duration. Unexpectantly, donepezil at 0.3 mg/kg CGP-35348 at 100 mg/kg had no effect on SWDs. These findings call for re-evaluation of some prevailing theories on neural circuit alternations that underlie SWD generation and show the utility of APP/PS1 mice for testing potential new treatments for nonconvulsive epileptic activity related to Alzheimer pathology.
Collapse
Affiliation(s)
- Nanxiang Jin
- A. I. Virtanen Institute, University of Eastern Finland, PO Box 1627, 70211, Kuopio, Finland.
| | - Sofya Ziyatdinova
- A. I. Virtanen Institute, University of Eastern Finland, PO Box 1627, 70211, Kuopio, Finland
| | - Irina Gureviciene
- A. I. Virtanen Institute, University of Eastern Finland, PO Box 1627, 70211, Kuopio, Finland
| | - Heikki Tanila
- A. I. Virtanen Institute, University of Eastern Finland, PO Box 1627, 70211, Kuopio, Finland
| |
Collapse
|
64
|
Epilepsy and Alzheimer’s Disease: Potential mechanisms for an association. Brain Res Bull 2020; 160:107-120. [DOI: 10.1016/j.brainresbull.2020.04.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/05/2020] [Accepted: 04/10/2020] [Indexed: 12/16/2022]
|
65
|
Casillas‐Espinosa PM, Ali I, O'Brien TJ. Neurodegenerative pathways as targets for acquired epilepsy therapy development. Epilepsia Open 2020; 5:138-154. [PMID: 32524040 PMCID: PMC7278567 DOI: 10.1002/epi4.12386] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/13/2020] [Accepted: 02/24/2020] [Indexed: 12/16/2022] Open
Abstract
There is a growing body of clinical and experimental evidence that neurodegenerative diseases and epileptogenesis after an acquired brain insult may share common etiological mechanisms. Acquired epilepsy commonly develops as a comorbid condition in patients with neurodegenerative diseases such as Alzheimer's disease, although it is likely much under diagnosed in practice. Progressive neurodegeneration has also been described after traumatic brain injury, stroke, and other forms of brain insults. Moreover, recent evidence has shown that acquired epilepsy is often a progressive disorder that is associated with the development of drug resistance, cognitive decline, and worsening of other neuropsychiatric comorbidities. Therefore, new pharmacological therapies that target neurobiological pathways that underpin neurodegenerative diseases have potential to have both an anti-epileptogenic and disease-modifying effect on the seizures in patients with acquired epilepsy, and also mitigate the progressive neurocognitive and neuropsychiatric comorbidities. Here, we review the neurodegenerative pathways that are plausible targets for the development of novel therapies that could prevent the development or modify the progression of acquired epilepsy, and the supporting published experimental and clinical evidence.
Collapse
Affiliation(s)
- Pablo M. Casillas‐Espinosa
- Departments of Neuroscience and MedicineCentral Clinical SchoolMonash UniversityMelbourneVic.Australia
- Department of MedicineThe Royal Melbourne HospitalThe University of MelbourneMelbourneVic.Australia
| | - Idrish Ali
- Departments of Neuroscience and MedicineCentral Clinical SchoolMonash UniversityMelbourneVic.Australia
- Department of MedicineThe Royal Melbourne HospitalThe University of MelbourneMelbourneVic.Australia
| | - Terence J. O'Brien
- Departments of Neuroscience and MedicineCentral Clinical SchoolMonash UniversityMelbourneVic.Australia
- Department of MedicineThe Royal Melbourne HospitalThe University of MelbourneMelbourneVic.Australia
- Department of NeurologyThe Alfred HospitalMelbourneVic.Australia
- Department of NeurologyThe Royal Melbourne HospitalParkvilleVic.Australia
| |
Collapse
|
66
|
Rizzello E, Middei S, Marchetti C. Synaptic Correlates of Anterograde Amnesia and Intact Retrograde Memory in a Mouse Model of Alzheimer’s Disease. Curr Alzheimer Res 2020; 17:259-268. [DOI: 10.2174/1567205017666200224122113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/30/2019] [Accepted: 02/12/2020] [Indexed: 01/08/2023]
Abstract
Background:
Clinical evidence indicates that patients affected by Alzheimer's Disease (AD)
fail to form new memories although their memories for old events are intact. This amnesic pattern depends
on the selective vulnerability to AD-neurodegeneration of the hippocampus, the brain region that
sustains the formation of new memories, while cortical regions that store remote memories are spared.
Objective:
To identify the cellular mechanisms underlying impaired recent memories and intact remote
memories in a mouse model of AD.
Method:
Glutamatergic synaptic currents were recorded by patch-clamp in acute hippocampal and
anterior Cingulate Cortical (aCC) slices of AD-like Tg2576 mice and Wild-type (Wt) littermates subjected
to the Contextual Fear Conditioning (CFC) task or in naïve conditions.
Results:
Glutamatergic synaptic currents were recorded by patch-clamp in acute hippocampal and
anterior Cingulate Cortical (aCC) slices of AD-like Tg2576 mice and Wild-type (Wt) littermates subjected
to the Contextual Fear Conditioning (CFC) task or in naïve conditions.
Conclusion:
Our data suggest that in the early AD stages synaptic plasticity of CA1 synapses, crucial
to form new memories, is lost, while plasticity of aCC synapses is intact and contributes to the persistence
of long-term memories.
Collapse
Affiliation(s)
- Emanuela Rizzello
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Via Regina Elena 295, 00161, Rome, Italy
| | - Silvia Middei
- Consiglio Nazionale delle Ricerche (CNR), Institute of Biochemistry and Cell Biology, Via E. Ramarini 32, 00015 Monterotondo, Rome, Italy
| | - Cristina Marchetti
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Via Regina Elena 295, 00161, Rome, Italy
| |
Collapse
|
67
|
Brunetti V, D'Atri A, Della Marca G, Vollono C, Marra C, Vita MG, Scarpelli S, De Gennaro L, Rossini PM. Subclinical epileptiform activity during sleep in Alzheimer's disease and mild cognitive impairment. Clin Neurophysiol 2020; 131:1011-1018. [PMID: 32193162 DOI: 10.1016/j.clinph.2020.02.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/23/2020] [Accepted: 02/06/2020] [Indexed: 02/08/2023]
|
68
|
Optimization and biological evaluation of imidazopyridine derivatives as a novel scaffold for γ-secretase modulators with oral efficacy against cognitive deficits in Alzheimer's disease model mice. Bioorg Med Chem 2020; 28:115455. [PMID: 32307259 DOI: 10.1016/j.bmc.2020.115455] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/17/2020] [Accepted: 03/20/2020] [Indexed: 01/18/2023]
Abstract
Gamma-secretase modulators (GSMs) selectively lower amyloid-β42 (Aβ42) and are therefore potential disease-modifying drugs for Alzheimer's disease (AD). Here, we report the discovery of imidazopyridine derivatives as GSMs with oral activity on not only Aβ42 levels but also cognitive function. Structural optimization of the biphenyl group and pyridine-2-amide moiety of compound 1a greatly improved GSM activity and rat microsomal stability, respectively. 5-{8-[(3,4'-Difluoro[1,1'-biphenyl]-4-yl)methoxy]-2-methylimidazo[1,2-a]pyridin-3-yl}-N-methylpyridine-2-carboxamide (1o) showed high in vitro potency and brain exposure, induced a robust reduction in brain Aβ42 levels, and exhibited undetectable inhibition of cytochrome p450 enzymes. Moreover, compound 1o showed excellent efficacy against cognitive deficits in AD model mice. These findings suggest that compound 1o is a promising candidate for AD therapeutics.
Collapse
|
69
|
Kam K, Kang M, Eren CY, Pettibone WD, Bowling H, Taveras S, Ly A, Chen RK, Berryman NV, Klann E, Varga AW. Interactions between sleep disruption, motor learning, and p70 S6 kinase 1 signaling. Sleep 2020; 43:zsz244. [PMID: 31608388 PMCID: PMC7315768 DOI: 10.1093/sleep/zsz244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 09/11/2019] [Indexed: 11/14/2022] Open
Abstract
Offline gains in motor performance after initial motor learning likely depend on sleep, but the molecular mechanisms by which this occurs are understudied. Regulation of mRNA translation via p70 S6 kinase 1 (S6K1) signaling represents one potential mechanism, as protein synthesis is thought to be increased during sleep compared to wake and is necessary for several forms of long-term memory. Using phosphorylation of ribosomal protein S6 (RpS6) as a readout of S6K1 activity, we demonstrate that a period of 10 h of acute sleep disruption impairs both S6K1 signaling and offline gains in motor performance on the rotarod in adult wild type C57/Bl6 mice. Rotarod motor learning results in increased abundance of RpS6 in the striatum, and inhibition of S6K1 either indirectly with rapamycin or directly with PF-4708671 diminished the offline improvement in motor performance without affecting the initial acquisition of rotarod motor learning when sleep is normal. In sum, S6K1 activity is required for sleep-dependent offline gains in motor performance and is inhibited following acute sleep disruption, while motor learning increases the abundance of striatal RpS6. Thus, S6K1 signaling represents a plausible mechanism mediating the beneficial effects of sleep on motor performance.
Collapse
Affiliation(s)
- Korey Kam
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Mihwa Kang
- Center for Neural Science, New York University, New York, NY
| | - C Yasemin Eren
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ward D Pettibone
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Heather Bowling
- Center for Neural Science, New York University, New York, NY
| | - Shantal Taveras
- Center for Neural Science, New York University, New York, NY
| | - Annie Ly
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rebecca K Chen
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Natasha V Berryman
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY
| | - Andrew W Varga
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
70
|
Yang J, Li Q. Manganese-Enhanced Magnetic Resonance Imaging: Application in Central Nervous System Diseases. Front Neurol 2020; 11:143. [PMID: 32161572 PMCID: PMC7052353 DOI: 10.3389/fneur.2020.00143] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/07/2020] [Indexed: 12/12/2022] Open
Abstract
Manganese-enhanced magnetic resonance imaging (MEMRI) relies on the strong paramagnetism of Mn2+. Mn2+ is a calcium ion analog and can enter excitable cells through voltage-gated calcium channels. Mn2+ can be transported along the axons of neurons via microtubule-based fast axonal transport. Based on these properties, MEMRI is used to describe neuroanatomical structures, monitor neural activity, and evaluate axonal transport rates. The application of MEMRI in preclinical animal models of central nervous system (CNS) diseases can provide more information for the study of disease mechanisms. In this article, we provide a brief review of MEMRI use in CNS diseases ranging from neurodegenerative diseases to brain injury and spinal cord injury.
Collapse
Affiliation(s)
- Jun Yang
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital & Cancer Center, Kunming, China
| | - Qinqing Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital & Cancer Center, Kunming, China
| |
Collapse
|
71
|
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
|
72
|
Senatorov VV, Friedman AR, Milikovsky DZ, Ofer J, Saar-Ashkenazy R, Charbash A, Jahan N, Chin G, Mihaly E, Lin JM, Ramsay HJ, Moghbel A, Preininger MK, Eddings CR, Harrison HV, Patel R, Shen Y, Ghanim H, Sheng H, Veksler R, Sudmant PH, Becker A, Hart B, Rogawski MA, Dillin A, Friedman A, Kaufer D. Blood-brain barrier dysfunction in aging induces hyperactivation of TGFβ signaling and chronic yet reversible neural dysfunction. Sci Transl Med 2019; 11:eaaw8283. [PMID: 31801886 DOI: 10.1126/scitranslmed.aaw8283] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/15/2019] [Accepted: 11/07/2019] [Indexed: 12/16/2022]
Abstract
Aging involves a decline in neural function that contributes to cognitive impairment and disease. However, the mechanisms underlying the transition from a young-and-healthy to aged-and-dysfunctional brain are not well understood. Here, we report breakdown of the vascular blood-brain barrier (BBB) in aging humans and rodents, which begins as early as middle age and progresses to the end of the life span. Gain-of-function and loss-of-function manipulations show that this BBB dysfunction triggers hyperactivation of transforming growth factor-β (TGFβ) signaling in astrocytes, which is necessary and sufficient to cause neural dysfunction and age-related pathology in rodents. Specifically, infusion of the serum protein albumin into the young rodent brain (mimicking BBB leakiness) induced astrocytic TGFβ signaling and an aged brain phenotype including aberrant electrocorticographic activity, vulnerability to seizures, and cognitive impairment. Furthermore, conditional genetic knockdown of astrocytic TGFβ receptors or pharmacological inhibition of TGFβ signaling reversed these symptomatic outcomes in aged mice. Last, we found that this same signaling pathway is activated in aging human subjects with BBB dysfunction. Our study identifies dysfunction in the neurovascular unit as one of the earliest triggers of neurological aging and demonstrates that the aging brain may retain considerable latent capacity, which can be revitalized by therapeutic inhibition of TGFβ signaling.
Collapse
Affiliation(s)
- Vladimir V Senatorov
- Helen Wills Neuroscience Institute and Berkeley Stem Cell Center, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Aaron R Friedman
- Helen Wills Neuroscience Institute and Berkeley Stem Cell Center, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Dan Z Milikovsky
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Jonathan Ofer
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Rotem Saar-Ashkenazy
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Adiel Charbash
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Naznin Jahan
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Gregory Chin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Eszter Mihaly
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jessica M Lin
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Harrison J Ramsay
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ariana Moghbel
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Marcela K Preininger
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chelsy R Eddings
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Helen V Harrison
- School of Public Health, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Rishi Patel
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yizhuo Shen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Hana Ghanim
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Huanjie Sheng
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ronel Veksler
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Peter H Sudmant
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Albert Becker
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany
| | - Barry Hart
- Innovation Pathways, Palo Alto, CA 94301, USA
| | - Michael A Rogawski
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Andrew Dillin
- Glenn Center for Aging Research, Howard Hughes Medical Institute, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
- Department of Medical Neuroscience and Brain Repair Center, Dalhousie University, Halifax, NS B3H4R2, Canada
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute and Berkeley Stem Cell Center, University of California, Berkeley, Berkeley, CA 94720, USA.
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Canadian Institute for Advanced Research, Toronto, ON M5G1M1, Canada
| |
Collapse
|
73
|
Gureviciene I, Ishchenko I, Ziyatdinova S, Jin N, Lipponen A, Gurevicius K, Tanila H. Characterization of Epileptic Spiking Associated With Brain Amyloidosis in APP/PS1 Mice. Front Neurol 2019; 10:1151. [PMID: 31781019 PMCID: PMC6861424 DOI: 10.3389/fneur.2019.01151] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 10/14/2019] [Indexed: 12/22/2022] Open
Abstract
Epileptic activity without visible convulsions is common in Alzheimer's disease (AD) and may contribute adversely to the disease progress and symptoms. Transgenic mice with amyloid plaque pathology also display epileptic seizures, but those are too infrequent to assess the effect of anti-epileptic treatments. Besides spontaneous seizures, these mice also display frequent epileptic spiking in epidural EEG recordings, and these have provided a means to test potential drug treatment to AD-related epilepsy. However, the origin of EEG spikes in transgenic AD model mice has remained elusive, which makes it difficult to relate electrophysiology with underlying pathology at the cellular and molecular level. Using multiple cortical and subcortical electrodes in freely moving APP/PS1 transgenic mice and their wild-type littermates, we identified several types of epileptic spikes among over 15 800 spikes visible with cortical screw electrodes based on their source localization. Cortical spikes associated with muscle twitches, cortico-hippocampal spikes, and spindle and fast-spindle associated spikes were present equally often in both APP/PS1 and wild-type mice, whereas pure cortical spikes were slightly more common in APP/PS1 mice. In contrast, spike-wave discharges, cortico-hippocampal spikes with after hyperpolarization and giant spikes were seen almost exclusively in APP/PS1 mice but only in a subset of them. Interestingly, different subtypes of spikes responded differently to anti-epileptic drugs ethosuximide and levetiracetam. From the translational point most relevant may be the giant spikes generated in the hippocampus that reached an amplitude up to ± 5 mV in the hippocampal channel. As in AD patients, they occurred exclusively during sleep. Further, we could demonstrate that a high number of giant spikes in APP/PS1 mice predicts seizures. These data show that by only adding a pair of hippocampal deep electrodes and EMG to routine cortical epidural screw electrodes and by taking into account underlying cortical oscillations, one can drastically refine the analysis of cortical spike data. This new approach provides a powerful tool to preclinical testing of potential new treatment options for AD related epilepsy.
Collapse
Affiliation(s)
- Irina Gureviciene
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Irina Ishchenko
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Sofya Ziyatdinova
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Nanxiang Jin
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Arto Lipponen
- Department of Psychology, University of Jyväskylä, Jyväskylä, Finland
| | | | - Heikki Tanila
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
74
|
Kam K, Pettibone WD, Shim K, Chen RK, Varga AW. Dynamics of sleep spindles and coupling to slow oscillations following motor learning in adult mice. Neurobiol Learn Mem 2019; 166:107100. [PMID: 31622665 DOI: 10.1016/j.nlm.2019.107100] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/18/2019] [Accepted: 10/11/2019] [Indexed: 01/05/2023]
Abstract
Sleep spindles have been implicated in motor learning in human subjects, but their occurrence, timing in relation to cortical slow oscillations, and relationship to offline gains in motor learning have not been examined in animal models. In this study, we recorded EEG over bilateral primary motor cortex in conjunction with EMG for 24 h following a period of either baseline handling or following rotarod motor learning to monitor sleep. We measured several biophysical properties of sleep spindles and their temporal coupling with cortical slow oscillations (SO, <1 Hz) and cortical delta waves (1-4 Hz). Following motor learning, we found an increase in spindles during an early period of NREM sleep (1-4 h) without changes to biophysical properties such as spindle power, peak frequency and coherence. In this same period of early NREM sleep, both SO and delta power increased after motor learning. Notably, a vast majority of spindles were associated with minimal SO power, but in the subset that were associated with significant SO power (>1 z-score above the population mean), spindle-associated SO power was greater in spindles following motor learning compared to baseline sleep. Also, we did not observe a group-level preferred phase in spindle-SO or spindle-delta coupling. While SO power alone was not predictive of motor performance in early NREM sleep, both spindle density and the difference in the magnitude of the mean resultant vector length of the phase angle for SO-associated spindles, a measure of its coupling precision, were positively correlated with offline change in motor performance. These findings support a role for sleep spindles and their coupling to slow oscillations in motor learning and establish a model in which spindle timing and the brain circuits that support offline plasticity can be mechanistically explored.
Collapse
Affiliation(s)
- Korey Kam
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ward D Pettibone
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kaitlyn Shim
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rebecca K Chen
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andrew W Varga
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
75
|
Ham HJ, Han SB, Yun J, Yeo IJ, Ham YW, Kim SH, Park PH, Choi DY, Hong JT. Bee venom phospholipase A2 ameliorates amyloidogenesis and neuroinflammation through inhibition of signal transducer and activator of transcription-3 pathway in Tg2576 mice. Transl Neurodegener 2019; 8:26. [PMID: 31592103 PMCID: PMC6774221 DOI: 10.1186/s40035-019-0167-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 08/12/2019] [Indexed: 12/21/2022] Open
Abstract
Background Neuroinflammation and accumulation of β-amyloid (Aβ) play a significant role in the onset and progression of Alzheimer’s disease (AD). Our previous study demonstrated that signal transducer and activator of transcription-3 (STAT3) plays a major role in neuroinflammation and amyloidogenesis. Methods In the present study, we investigated the inhibitory effect of bee venom phospholipase A2 (bvPLA2) on memory deficiency in Tg2576 mice, which demonstrate genetic characteristics of AD and the mechanism of its action at the cellular and animal level. For in vivo study, we examined the effect of bvPLA2 on improving memory by conducting several behavioral tests with the administration of bvPLA2 (1 mg/kg) to Tg2576 mice. For in vitro study, we examined the effect of bvPLA2 on amyloidogenesis and neuroinflammation by treating bvPLA2 on LPS-activated BV2 cells. Results We found that bvPLA2 alleviated memory impairment in Tg2576 mice, as demonstrated in the behavioral tests assessing memory. In the bvPLA2-treated group, Aβ, amyloid precursor protein (APP), and β-secretase 1 (BACE1) levels and β-secretase activity were significantly decreased. Expression of pro-inflammatory cytokines and inflammation-related proteins decreased in the brain of bvPLA2-treated group, whereas anti-inflammatory cytokines increased. In addition, bvPLA2 reduced STAT3 phosphorylation in the brains of the bvPLA2-treated group. At the cellular level, bvPLA2 inhibits production of nitric oxide, pro-inflammatory cytokines, and inflammation-related proteins including p-STAT3. Additionally, bvPLA2 inhibits the production of Aβ in cultured BV-2 cells. Results from the docking experiment, pull-down assay, and the luciferase assay show that bvPLA2 directly binds STAT3 and, thus, regulates gene expression levels. Moreover, when the STAT3 inhibitor and bvPLA2 were administered together, the anti-amyloidogenic and anti-inflammatory effects were further enhanced than when they were administered alone. Conclusion These results suggest that bvPLA2 could restore memory by inhibiting the accumulation of Aβ and inflammatory responses via blockage of STAT3 activity. Electronic supplementary material The online version of this article (10.1186/s40035-019-0167-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hyeon Joo Ham
- 1College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160 Republic of Korea
| | - Sang-Bae Han
- 1College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160 Republic of Korea
| | - Jaesuk Yun
- 1College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160 Republic of Korea
| | - In Jun Yeo
- 1College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160 Republic of Korea
| | - Young Wan Ham
- 2Department of Chemistry, Utah Valley University, 800 W University Pkwy, Orem, UT 84058 USA
| | - Se Hyun Kim
- INISTst Co., LTD, 767, Sinsu-ro, Suji-gu, Yongin-si, 16827 Gyeonggi-do Republic of Korea
| | - Pil-Hoon Park
- 4College of Pharmacy, Yeungnam University, 280 Daehak Road, Gyeonsan, Gyeongbuk, 38541 Republic of Korea
| | - Dong-Young Choi
- 4College of Pharmacy, Yeungnam University, 280 Daehak Road, Gyeonsan, Gyeongbuk, 38541 Republic of Korea
| | - Jin Tae Hong
- 1College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160 Republic of Korea
| |
Collapse
|
76
|
Carvalho DZ, St Louis EK, Knopman DS, Boeve BF, Lowe VJ, Roberts RO, Mielke MM, Przybelski SA, Machulda MM, Petersen RC, Jack CR, Vemuri P. Association of Excessive Daytime Sleepiness With Longitudinal β-Amyloid Accumulation in Elderly Persons Without Dementia. JAMA Neurol 2019. [PMID: 29532057 DOI: 10.1001/jamaneurol.2018.0049] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Importance Aging is associated with excessive daytime sleepiness (EDS), which has been linked to cognitive decline in the elderly. However, whether EDS is associated with the pathologic processes of Alzheimer disease remains unclear. Objective To investigate whether EDS at baseline is associated with a longitudinal increase in regional β-amyloid (Aβ) accumulation in a cohort of elderly individuals without dementia. Design, Setting, and Participants This prospective analysis included participants enrolled in the Mayo Clinic Study of Aging, a longitudinal population-based study in Olmsted County, Minnesota. Of 2900 participants, 2172 (74.9%) agreed to undergo carbon 11-labeled Pittsburgh compound B positron emission tomography (PiB-PET). We included 283 participants 70 years or older without dementia who completed surveys assessing sleepiness at baseline and had at least 2 consecutive PiB-PET scans from January 1, 2009, through July 31, 2016, after excluding 45 (13.7%) who had a comorbid neurologic disorder. Main Outcomes and Measures Excessive daytime sleepiness was defined as an Epworth Sleepiness Scale score of at least 10. The difference in Aβ levels between the 2 consecutive scans (ΔPiB) in Aβ-susceptible regions (prefrontal, anterior cingulate, posterior cingulate-precuneus, and parietal) was determined. Multiple linear regression models were fit to explore associations between baseline EDS and ΔPiB while adjusting for baseline age, sex, presence of the apolipoprotein E ε4 allele, educational level, baseline PiB uptake, global PiB positivity (standardized uptake value ratio ≥1.4), physical activity, cardiovascular comorbidities (obesity, hypertension, hyperlipidemia, and diabetes), reduced sleep duration, respiratory symptoms during sleep, depression, and interval between scans. Results Of the initial 283 participants, mean (SD) age was 77.1 (4.8) years; 204 (72.1%) were men and 79 (27.9%) were women. Sixty-three participants (22.3%) had EDS. Baseline EDS was significantly associated with increased regional Aβ accumulation in the anterior cingulate (B coefficient = 0.031; 95% CI, 0.001-0.061; P = .04), posterior cingulate-precuneus (B coefficient = 0.038; 95% CI, 0.006-0.069; P = .02), and parietal (B coefficient = 0.033; 95% CI, 0.001-0.065; P = .04) regions. Association of EDS with longitudinal Aβ accumulation was stronger in participants with baseline global PiB positivity in the anterior cingulate (B coefficient = 0.065; 95% CI, 0.010-0.118; P = .02) and cingulate-precuneus (B coefficient = 0.068; 95% CI, 0.009-0.126; P = .02) regions. Conclusions and Relevance Baseline EDS was associated with increased longitudinal Aβ accumulation in elderly persons without dementia, suggesting that those with EDS may be more vulnerable to pathologic changes associated with Alzheimer disease. Further work is needed to elucidate whether EDS is a clinical marker of greater sleep instability, synaptic or network overload, or neurodegeneration of wakefulness-promoting centers. Early identification of patients with EDS and treatment of underlying sleep disorders could reduce Aβ accumulation in this vulnerable group.
Collapse
Affiliation(s)
| | | | | | | | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Rosebud O Roberts
- Department of Neurology, Mayo Clinic, Rochester, Minnesota.,Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Michelle M Mielke
- Department of Neurology, Mayo Clinic, Rochester, Minnesota.,Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Scott A Przybelski
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | | | | | | | | |
Collapse
|
77
|
Varma P, Hsieh J. Rise and Fall of the Empire: Conquering Alzheimer's Disease by Targeting Adult Neurogenesis. Epilepsy Curr 2019; 19:411-413. [PMID: 31526033 PMCID: PMC6891180 DOI: 10.1177/1535759719874813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
78
|
Amyloid β-Induced Upregulation of Na v1.6 Underlies Neuronal Hyperactivity in Tg2576 Alzheimer's Disease Mouse Model. Sci Rep 2019; 9:13592. [PMID: 31537873 PMCID: PMC6753212 DOI: 10.1038/s41598-019-50018-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 07/31/2019] [Indexed: 12/19/2022] Open
Abstract
Hyperexcitability and alterations in neuronal networks contribute to cognitive impairment in Alzheimer’s Disease (AD). Voltage-gated sodium channels (NaV), which are crucial for regulating neuronal excitability, have been implicated in AD-related hippocampal hyperactivity and higher incidence of spontaneous non-convulsive seizures. Here, we show by using primary hippocampal neurons exposed to amyloid-β1–42 (Aβ1–42) oligomers and from Tg2576 mouse embryos, that the selective upregulation of NaV1.6 subtype contributes to membrane depolarization and to the increase of spike frequency, thereby resulting in neuronal hyperexcitability. Interestingly, we also found that NaV1.6 overexpression is responsible for the aberrant neuronal activity observed in hippocampal slices from 3-month-old Tg2576 mice. These findings identify the NaV1.6 channels as a determinant of the hippocampal neuronal hyperexcitability induced by Aβ1–42 oligomers. The selective blockade of NaV1.6 overexpression and/or hyperactivity might therefore offer a new potential therapeutic approach to counteract early hippocampal hyperexcitability and subsequent cognitive deficits in the early stages of AD.
Collapse
|
79
|
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
|
80
|
Fu CH, Iascone DM, Petrof I, Hazra A, Zhang X, Pyfer MS, Tosi U, Corbett BF, Cai J, Lee J, Park J, Iacovitti L, Scharfman HE, Enikolopov G, Chin J. Early Seizure Activity Accelerates Depletion of Hippocampal Neural Stem Cells and Impairs Spatial Discrimination in an Alzheimer's Disease Model. Cell Rep 2019; 27:3741-3751.e4. [PMID: 31242408 PMCID: PMC6697001 DOI: 10.1016/j.celrep.2019.05.101] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 04/24/2019] [Accepted: 05/28/2019] [Indexed: 12/21/2022] Open
Abstract
Adult hippocampal neurogenesis has been reported to be decreased, increased, or not changed in Alzheimer's disease (AD) patients and related transgenic mouse models. These disparate findings may relate to differences in disease stage, or the presence of seizures, which are associated with AD and can stimulate neurogenesis. In this study, we investigate a transgenic mouse model of AD that exhibits seizures similarly to AD patients and find that neurogenesis is increased in early stages of disease, as spontaneous seizures became evident, but is decreased below control levels as seizures recur. Treatment with the antiseizure drug levetiracetam restores neurogenesis and improves performance in a neurogenesis-associated spatial discrimination task. Our results suggest that seizures stimulate, and later accelerate the depletion of, the hippocampal neural stem cell pool. These results have implications for AD as well as any disorder accompanied by recurrent seizures, such as epilepsy.
Collapse
Affiliation(s)
- Chia-Hsuan Fu
- Memory & Brain Research Center, Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Daniel Maxim Iascone
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Iraklis Petrof
- Memory & Brain Research Center, Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Anupam Hazra
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Xiaohong Zhang
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Mark S Pyfer
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Umberto Tosi
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Brian F Corbett
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jingli Cai
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jason Lee
- Memory & Brain Research Center, Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jin Park
- Memory & Brain Research Center, Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lorraine Iacovitti
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Helen E Scharfman
- Departments of Psychiatry, Neuroscience, and Physiology and the Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Grigori Enikolopov
- Center for Developmental Genetics and Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jeannie Chin
- Memory & Brain Research Center, Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
81
|
Kulkarni PM, Xiao Z, Robinson EJ, Jami AS, Zhang J, Zhou H, Henin SE, Liu AA, Osorio RS, Wang J, Chen Z. A deep learning approach for real-time detection of sleep spindles. J Neural Eng 2019; 16:036004. [PMID: 30790769 PMCID: PMC6527330 DOI: 10.1088/1741-2552/ab0933] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Sleep spindles have been implicated in memory consolidation and synaptic plasticity during NREM sleep. Detection accuracy and latency in automatic spindle detection are critical for real-time applications. APPROACH Here we propose a novel deep learning strategy (SpindleNet) to detect sleep spindles based on a single EEG channel. While the majority of spindle detection methods are used for off-line applications, our method is well suited for online applications. MAIN RESULTS Compared with other spindle detection methods, SpindleNet achieves superior detection accuracy and speed, as demonstrated in two publicly available expert-validated EEG sleep spindle datasets. Our real-time detection of spindle onset achieves detection latencies of 150-350 ms (~two-three spindle cycles) and retains excellent performance under low EEG sampling frequencies and low signal-to-noise ratios. SpindleNet has good generalization across different sleep datasets from various subject groups of different ages and species. SIGNIFICANCE SpindleNet is ultra-fast and scalable to multichannel EEG recordings, with an accuracy level comparable to human experts, making it appealing for long-term sleep monitoring and closed-loop neuroscience experiments.
Collapse
Affiliation(s)
- Prathamesh M Kulkarni
- Department of Psychiatry, School of Medicine, New York University, New York, NY 10016, United States of America
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
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
|
83
|
Pettibone WD, Kam K, Chen RK, Varga AW. Necessity of Sleep for Motor Gist Learning in Mice. Front Neurosci 2019; 13:293. [PMID: 31024231 PMCID: PMC6459967 DOI: 10.3389/fnins.2019.00293] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/13/2019] [Indexed: 12/20/2022] Open
Abstract
With respect to behavior, the term memory "consolidation" has canonically been used to describe increased fidelity during testing to a learned behavior shaped during training. While the sleeping brain appears to certainly aid in consolidation by this definition for a variety of memories, including motor memories, growing evidence suggests that sleep allows for much more flexible use of the information encountered during prior wakefulness. Sleep has been shown to augment the extraction of gist or patterns from wake experience in human subjects, but this has been difficult to recapitulate in animal models owing to the semantic requirements in many such tasks. Here we establish a model of motor gist learning in mice in which two bouts of exclusive forward running on the rotarod significantly augments the first experience of exclusive backward running. This augmentation does not occur if sleep is disrupted following the forward running template behavior or if a period of natural wakefulness follows one of the two bouts of exclusive forward running. This suggests that sleep is required for the extraction of the motor gist of forward running to apply to backward running.
Collapse
Affiliation(s)
| | | | | | - Andrew W. Varga
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
84
|
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
|
85
|
Saar G, Koretsky AP. Manganese Enhanced MRI for Use in Studying Neurodegenerative Diseases. Front Neural Circuits 2019; 12:114. [PMID: 30666190 PMCID: PMC6330305 DOI: 10.3389/fncir.2018.00114] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 12/10/2018] [Indexed: 12/13/2022] Open
Abstract
MRI has been extensively used in neurodegenerative disorders, such as Alzheimer’s disease (AD), frontal-temporal dementia (FTD), mild cognitive impairment (MCI), Parkinson’s disease (PD), Huntington’s disease (HD) and amyotrophic lateral sclerosis (ALS). MRI is important for monitoring the neurodegenerative components in other diseases such as epilepsy, stroke and multiple sclerosis (MS). Manganese enhanced MRI (MEMRI) has been used in many preclinical studies to image anatomy and cytoarchitecture, to obtain functional information in areas of the brain and to study neuronal connections. This is due to Mn2+ ability to enter excitable cells through voltage gated calcium channels and be actively transported in an anterograde manner along axons and across synapses. The broad range of information obtained from MEMRI has led to the use of Mn2+ in many animal models of neurodegeneration which has supplied important insight into brain degeneration in preclinical studies. Here we provide a brief review of MEMRI use in neurodegenerative diseases and in diseases with neurodegenerative components in animal studies and discuss the potential translation of MEMRI to clinical use in the future.
Collapse
Affiliation(s)
- Galit Saar
- Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD, United States
| | - Alan P Koretsky
- Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
86
|
Cattaud V, Bezzina C, Rey CC, Lejards C, Dahan L, Verret L. Early disruption of parvalbumin expression and perineuronal nets in the hippocampus of the Tg2576 mouse model of Alzheimer's disease can be rescued by enriched environment. Neurobiol Aging 2018; 72:147-158. [DOI: 10.1016/j.neurobiolaging.2018.08.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/28/2018] [Accepted: 08/28/2018] [Indexed: 11/30/2022]
|
87
|
Liu D, Lu H, Stein E, Zhou Z, Yang Y, Mattson MP. Brain regional synchronous activity predicts tauopathy in 3×TgAD mice. Neurobiol Aging 2018; 70:160-169. [PMID: 30015035 DOI: 10.1016/j.neurobiolaging.2018.06.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/20/2018] [Accepted: 06/10/2018] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is characterized by progressive cognitive impairment and by extensive neuronal loss associated with extracellular amyloid β-peptide (Aβ) plaques and intraneuronal tau pathology in temporal and parietal lobes. AD patients are at increased risk for epileptic seizures, and data from experimental models of AD suggest that aberrant neuronal network activity occurs early in the disease process before cognitive deficits and neuronal degeneration. The contributions of Aβ and/or tau pathologies to dysregulation of neuronal network activity are unclear. Using a transgenic mouse model of AD (3×TgAD mice) in which there occurs differential age-dependent development of tau and Aβ plaque pathologies, we applied analysis of resting state functional magnetic resonance imaging regional homogeneity, a measure of local synchronous activity, to discriminate the effects of Aβ and tau on neuronal network activity throughout the brain. Compared to age-matched wild-type mice, 6- to 8-month-old 3×TgAD mice exhibited increased regional homogeneity in the hippocampus and parietal and temporal cortices, regions with tau pathology but not Aβ pathology at this age. By 18-24 months of age, 3×TgAD mice exhibited extensive tau and Aβ pathologies involving the hippocampus and multiple functionally related brain regions, with a spatial expansion of increased local synchronous activity to include those regions. Our findings demonstrate that age-related brain regional hypersynchronous activity is associated with early tau pathology in a mouse model, consistent with a role for early tau pathology in the neuronal circuit hyperexcitability that is believed to precede and contribute to neuronal degeneration in AD.
Collapse
Affiliation(s)
- Dong Liu
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, USA
| | - Hanbing Lu
- Neuroimaging Research Branch, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, USA
| | - Elliot Stein
- Neuroimaging Research Branch, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, USA
| | - Zhujuan Zhou
- Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yihong Yang
- Neuroimaging Research Branch, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, USA
| | - Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore MD, USA.
| |
Collapse
|
88
|
Gschwind T, Lafourcade C, Gfeller T, Zaichuk M, Rambousek L, Knuesel I, Fritschy JM. Contribution of early Alzheimer's disease-related pathophysiology to the development of acquired epilepsy. Eur J Neurosci 2018; 47:1534-1562. [DOI: 10.1111/ejn.13983] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 04/25/2018] [Accepted: 05/29/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Tilo Gschwind
- Institute of Pharmacology and Toxicology; University of Zurich; Zurich Switzerland
- Neuroscience Center Zurich; University of Zurich and ETH Zurich; Zurich Switzerland
| | - Carlos Lafourcade
- Institute of Pharmacology and Toxicology; University of Zurich; Zurich Switzerland
- Laboratorio de Neurociencias; Universidad de los Andes; Santiago Chile
| | - Tim Gfeller
- Institute of Pharmacology and Toxicology; University of Zurich; Zurich Switzerland
| | - Mariana Zaichuk
- Institute of Pharmacology and Toxicology; University of Zurich; Zurich Switzerland
- Neuroscience Center Zurich; University of Zurich and ETH Zurich; Zurich Switzerland
| | - Lukas Rambousek
- Institute of Experimental Immunology; University of Zurich; Zurich Switzerland
| | - Irene Knuesel
- Institute of Pharmacology and Toxicology; University of Zurich; Zurich Switzerland
- Roche Pharmaceutical Research and Early Development; NORD Discovery & Translational Area; Roche Innovation Center Basel; Basel Switzerland
| | - Jean-Marc Fritschy
- Institute of Pharmacology and Toxicology; University of Zurich; Zurich Switzerland
- Neuroscience Center Zurich; University of Zurich and ETH Zurich; Zurich Switzerland
| |
Collapse
|
89
|
Neuronal levels and sequence of tau modulate the power of brain rhythms. Neurobiol Dis 2018; 117:181-188. [PMID: 29859869 DOI: 10.1016/j.nbd.2018.05.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/23/2018] [Accepted: 05/30/2018] [Indexed: 01/15/2023] Open
Abstract
Neural network dysfunction may contribute to functional decline and disease progression in neurodegenerative disorders. Diverse lines of evidence suggest that neuronal accumulation of tau promotes network dysfunction and cognitive decline. The A152T-variant of human tau (hTau-A152T) increases the risk of Alzheimer's disease (AD) and several other tauopathies. When overexpressed in neurons of transgenic mice, it causes age-dependent neuronal loss and cognitive decline, as well as non-convulsive epileptic activity, which is also seen in patients with AD. Using intracranial EEG recordings with electrodes implanted over the parietal cortex, we demonstrate that hTau-A152T increases the power of brain oscillations in the 0.5-6 Hz range more than wildtype human tau in transgenic lines with comparable levels of human tau protein in brain, and that genetic ablation of endogenous tau in Mapt-/- mice decreases the power of these oscillations as compared to wildtype controls. Suppression of hTau-A152T production in doxycycline-regulatable transgenic mice reversed their abnormal network activity. Treatment of hTau-A152T mice with the antiepileptic drug levetiracetam also rapidly and persistently reversed their brain dysrhythmia and network hypersynchrony. These findings suggest that both the level and the sequence of tau modulate the power of specific brain oscillations. The potential of EEG spectral changes as a biomarker deserves to be explored in clinical trials of tau-lowering therapeutics. Our results also suggest that levetiracetam treatment is able to counteract tau-dependent neural network dysfunction. Tau reduction and levetiracetam treatment may be of benefit in AD and other conditions associated with brain dysrhythmias and network hypersynchrony.
Collapse
|
90
|
Circadian and Brain State Modulation of Network Hyperexcitability in Alzheimer's Disease. eNeuro 2018; 5:eN-CFN-0426-17. [PMID: 29780880 PMCID: PMC5956746 DOI: 10.1523/eneuro.0426-17.2018] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/08/2018] [Accepted: 04/06/2018] [Indexed: 01/08/2023] Open
Abstract
Network hyperexcitability is a feature of Alzheimer' disease (AD) as well as numerous transgenic mouse models of AD. While hyperexcitability in AD patients and AD animal models share certain features, the mechanistic overlap remains to be established. We aimed to identify features of network hyperexcitability in AD models that can be related to epileptiform activity signatures in AD patients. We studied network hyperexcitability in mice expressing amyloid precursor protein (APP) with mutations that cause familial AD, and compared a transgenic model that overexpresses human APP (hAPP) (J20), to a knock-in model expressing APP at physiological levels (APPNL/F). We recorded continuous long-term electrocorticogram (ECoG) activity from mice, and studied modulation by circadian cycle, behavioral, and brain state. We report that while J20s exhibit frequent interictal spikes (IISs), APPNL/F mice do not. In J20 mice, IISs were most prevalent during daylight hours and the circadian modulation was associated with sleep. Further analysis of brain state revealed that IIS in J20s are associated with features of rapid eye movement (REM) sleep. We found no evidence of cholinergic changes that may contribute to IIS-circadian coupling in J20s. In contrast to J20s, intracranial recordings capturing IIS in AD patients demonstrated frequent IIS in non-REM (NREM) sleep. The salient differences in sleep-stage coupling of IIS in APP overexpressing mice and AD patients suggests that different mechanisms may underlie network hyperexcitability in mice and humans. We posit that sleep-stage coupling of IIS should be an important consideration in identifying mouse AD models that most closely recapitulate network hyperexcitability in human AD.
Collapse
|
91
|
Corbett BF, You JC, Zhang X, Pyfer MS, Tosi U, Iascone DM, Petrof I, Hazra A, Fu CH, Stephens GS, Ashok AA, Aschmies S, Zhao L, Nestler EJ, Chin J. ΔFosB Regulates Gene Expression and Cognitive Dysfunction in a Mouse Model of Alzheimer's Disease. Cell Rep 2018; 20:344-355. [PMID: 28700937 DOI: 10.1016/j.celrep.2017.06.040] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 03/15/2017] [Accepted: 06/15/2017] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by cognitive decline and 5- to 10-fold increased seizure incidence. How seizures contribute to cognitive decline in AD or other disorders is unclear. We show that spontaneous seizures increase expression of ΔFosB, a highly stable Fos-family transcription factor, in the hippocampus of an AD mouse model. ΔFosB suppressed expression of the immediate early gene c-Fos, which is critical for plasticity and cognition, by binding its promoter and triggering histone deacetylation. Acute histone deacetylase (HDAC) inhibition or inhibition of ΔFosB activity restored c-Fos induction and improved cognition in AD mice. Administration of seizure-inducing agents to nontransgenic mice also resulted in ΔFosB-mediated suppression of c-Fos, suggesting that this mechanism is not confined to AD mice. These results explain observations that c-Fos expression increases after acute neuronal activity but decreases with chronic activity. Moreover, these results indicate a general mechanism by which seizures contribute to persistent cognitive deficits, even during seizure-free periods.
Collapse
Affiliation(s)
- Brian F Corbett
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jason C You
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Xiaohong Zhang
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Mark S Pyfer
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Umberto Tosi
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Daniel M Iascone
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Iraklis Petrof
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Anupam Hazra
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Chia-Hsuan Fu
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Gabriel S Stephens
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Annie A Ashok
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Suzan Aschmies
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Lijuan Zhao
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Eric J Nestler
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jeannie Chin
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
92
|
Scharfman HE, Kanner AM, Friedman A, Blümcke I, Crocker CE, Cendes F, Diaz-Arrastia R, Förstl H, Fenton AA, Grace AA, Palop J, Morrison J, Nehlig A, Prasad A, Wilcox KS, Jette N, Pohlmann-Eden B. Epilepsy as a Network Disorder (2): What can we learn from other network disorders such as dementia and schizophrenia, and what are the implications for translational research? Epilepsy Behav 2018; 78:302-312. [PMID: 29097123 PMCID: PMC5756681 DOI: 10.1016/j.yebeh.2017.09.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 09/18/2017] [Accepted: 09/18/2017] [Indexed: 12/18/2022]
Abstract
There is common agreement that many disorders of the central nervous system are 'complex', that is, there are many potential factors that influence the development of the disease, underlying mechanisms, and successful treatment. Most of these disorders, unfortunately, have no cure at the present time, and therapeutic strategies often have debilitating side effects. Interestingly, some of the 'complexities' of one disorder are found in another, and the similarities are often network defects. It seems likely that more discussions of these commonalities could advance our understanding and, therefore, have clinical implications or translational impact. With this in mind, the Fourth International Halifax Epilepsy Conference and Retreat was held as described in the prior paper, and this companion paper focuses on the second half of the meeting. Leaders in various subspecialties of epilepsy research were asked to address aging and dementia or psychosis in people with epilepsy (PWE). Commonalities between autism, depression, aging and dementia, psychosis, and epilepsy were the focus of the presentations and discussion. In the last session, additional experts commented on new conceptualization of translational epilepsy research efforts. Here, the presentations are reviewed, and salient points are highlighted.
Collapse
Affiliation(s)
- Helen E Scharfman
- Departments of Psychiatry, Neurosciences and Physiology, and the Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA.
| | - Andres M Kanner
- University of Miami, Miller School of Medicine, 1120 NW 14th Street, Room #1324, Miami, FL 33136, USA
| | - Alon Friedman
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada; Department of Pediatrics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada; Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ingmar Blümcke
- Neuropathological Institute, University Hospitals Erlangen, Germany
| | - Candice E Crocker
- Nova Scotia Early Psychosis Program, Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Fernando Cendes
- Department of Neurology, University of Campinas, 13083-888 Campinas, Sao Paulo, Brazil
| | - Ramon Diaz-Arrastia
- Centre for Neuroscience & Regenerative Medicine, Uniformed Services University of the Health Sciences, 12725 Twinbrook Parkway, Rockville, MD 20852, USA
| | - Hans Förstl
- Department of Psychiatry, University of Munich, Klinikum rechts der Isar, Ismaninger Strabe 22, D-81675 Munich, Germany
| | - André A Fenton
- Centre for Neural Science, New York University, 4 Washington Place, Room 809, New York, NY 10003, USA
| | - Anthony A Grace
- University of Pittsburgh, 456 Langley Hall, 4200 Fifth Avenue, Pittsburgh, PA 15269, USA
| | - Jorge Palop
- Department of Neurology, Gladstone Institute, 1650 Owens Street, San Francisco, CA 94158-2261, USA
| | - Jason Morrison
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Astrid Nehlig
- INSERM U 1129, Hôpital Necker, Paris, Faculty of Medicine, Strasbourg, France
| | - Asuri Prasad
- Department of Pediatrics, Children's Hospital of Western Ontario, London, ON, Canada
| | - Karen S Wilcox
- Department of Pharmacology & Toxicology, Anticonvulsant Drug Development Program, University of Utah, Salt Lake City, UT, USA
| | - Nathalie Jette
- Icahn School of Medicine at Mount Sinai, Department of Neurology, New York, NY, USA; Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Bernd Pohlmann-Eden
- Brain Repair Center, Life Science Research Institute, Dalhousie University, Room 229, PO Box 15000, Halifax, NS B3H4R2, Canada.
| |
Collapse
|
93
|
Vossel KA, Tartaglia MC, Nygaard HB, Zeman AZ, Miller BL. Epileptic activity in Alzheimer's disease: causes and clinical relevance. Lancet Neurol 2017; 16:311-322. [PMID: 28327340 DOI: 10.1016/s1474-4422(17)30044-3] [Citation(s) in RCA: 359] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 01/10/2017] [Accepted: 01/31/2017] [Indexed: 01/18/2023]
Abstract
Epileptic activity is frequently associated with Alzheimer's disease; this association has therapeutic implications, because epileptic activity can occur at early disease stages and might contribute to pathogenesis. In clinical practice, seizures in patients with Alzheimer's disease can easily go unrecognised because they usually present as non-motor seizures, and can overlap with other symptoms of the disease. In patients with Alzheimer's disease, seizures can hasten cognitive decline, highlighting the clinical relevance of early recognition and treatment. Some evidence indicates that subclinical epileptiform activity in patients with Alzheimer's disease, detected by extended neurophysiological monitoring, can also lead to accelerated cognitive decline. Treatment of clinical seizures in patients with Alzheimer's disease with select antiepileptic drugs (AEDs), in low doses, is usually well tolerated and efficacious. Moreover, studies in mouse models of Alzheimer's disease suggest that certain classes of AEDs that reduce network hyperexcitability have disease-modifying properties. These AEDs target mechanisms of epileptogenesis involving amyloid β and tau. Clinical trials targeting network hyperexcitability in patients with Alzheimer's disease will identify whether AEDs or related strategies could improve their cognitive symptoms or slow decline.
Collapse
Affiliation(s)
- Keith A Vossel
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA.
| | | | - Haakon B Nygaard
- Division of Neurology, University of British Columbia, Vancouver, BC, Canada
| | - Adam Z Zeman
- Cognitive Neurology Research Group, University of Exeter Medical School, Exeter, UK
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| |
Collapse
|
94
|
Lam AD, Deck G, Goldman A, Eskandar EN, Noebels J, Cole AJ. Silent hippocampal seizures and spikes identified by foramen ovale electrodes in Alzheimer's disease. Nat Med 2017; 23:678-680. [PMID: 28459436 PMCID: PMC5461182 DOI: 10.1038/nm.4330] [Citation(s) in RCA: 255] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 03/17/2017] [Indexed: 01/08/2023]
Abstract
We directly assessed mesial temporal activity in two Alzheimer’s disease (AD) patients without a history or EEG evidence of seizures, using intracranial foramen ovale electrodes. We detected clinically silent hippocampal seizures and epileptiform spikes during sleep, a period when both were most likely to interfere with memory consolidation. These index cases support a model in which early development of occult hippocampal hyperexcitability may contribute to the pathogenesis of AD.
Collapse
Affiliation(s)
- Alice D Lam
- MGH Epilepsy Service, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gina Deck
- MGH Epilepsy Service, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Alica Goldman
- Department of Neurology, Baylor College of Medicine, Houston, Texas, USA
| | - Emad N Eskandar
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jeffrey Noebels
- Department of Neurology, Baylor College of Medicine, Houston, Texas, USA
| | - Andrew J Cole
- MGH Epilepsy Service, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
95
|
Kazim SF, Chuang SC, Zhao W, Wong RKS, Bianchi R, Iqbal K. Early-Onset Network Hyperexcitability in Presymptomatic Alzheimer's Disease Transgenic Mice Is Suppressed by Passive Immunization with Anti-Human APP/Aβ Antibody and by mGluR5 Blockade. Front Aging Neurosci 2017; 9:71. [PMID: 28392767 PMCID: PMC5364175 DOI: 10.3389/fnagi.2017.00071] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 03/08/2017] [Indexed: 11/26/2022] Open
Abstract
Cortical and hippocampal network hyperexcitability appears to be an early event in Alzheimer’s disease (AD) pathogenesis, and may contribute to memory impairment. It remains unclear if network hyperexcitability precedes memory impairment in mouse models of AD and what are the underlying cellular mechanisms. We thus evaluated seizure susceptibility and hippocampal network hyperexcitability at ~3 weeks of age [prior to amyloid beta (Aβ) plaque deposition, neurofibrillary pathology, and cognitive impairment] in a triple transgenic mouse model of familial AD (3xTg-AD mouse) that harbors mutated human Aβ precursor protein (APP), tau and presenilin 1 (PS1) genes. Audiogenic seizures were elicited in a higher proportion of 3xTg-AD mice compared with wild type (WT) controls. Seizure susceptibility in 3xTg-AD mice was attenuated either by passive immunization with anti-human APP/Aβ antibody (6E10) or by blockade of metabotropic glutamate receptor 5 (mGluR5) with the selective antagonist, 2-methyl-6-(phenylethynyl)pyridine hydrochloride (MPEP). In in vitro hippocampal slices, suppression of synaptic inhibition with the GABAA receptor antagonist, bicuculline, induced prolonged epileptiform (>1.5 s in duration) ictal-like discharges in the CA3 neuronal network in the majority of the slices from 3xTg-AD mice. In contrast, only short epileptiform (<1.5 s in duration) interictal-like discharges were observed following bicuculline application in the CA3 region of WT slices. The ictal-like activity in CA3 region of the hippocampus was significantly reduced in the 6E10-immunized compared to the saline-treated 3xTg-AD mice. MPEP acutely suppressed the ictal-like discharges in 3xTg-AD slices. Remarkably, epileptiform discharge duration positively correlated with intraneuronal human (transgenic) APP/Aβ expression in the CA3 region of the hippocampus. Our data suggest that in a mouse model of familial AD, hypersynchronous network activity underlying seizure susceptibility precedes Aβ plaque pathology and memory impairment. This early-onset network hyperexcitability can be suppressed by passive immunization with an anti-human APP/Aβ antibody and by mGluR5 blockade in 3xTg-AD mice.
Collapse
Affiliation(s)
- Syed F Kazim
- Robert F. Furchgott Center for Neural and Behavioral Science and Department of Physiology and Pharmacology, State University of New York (SUNY) Downstate Medical CenterBrooklyn, NY, USA; Department of Neurochemistry and SUNY Downstate/NYSIBR Center for Developmental Neuroscience, New York State Institute for Basic Research (NYSIBR)Staten Island, NY, USA; Graduate Program in Neural and Behavioral Science, SUNY Downstate Medical CenterBrooklyn, NY, USA
| | - Shih-Chieh Chuang
- Robert F. Furchgott Center for Neural and Behavioral Science and Department of Physiology and Pharmacology, State University of New York (SUNY) Downstate Medical Center Brooklyn, NY, USA
| | - Wangfa Zhao
- Robert F. Furchgott Center for Neural and Behavioral Science and Department of Physiology and Pharmacology, State University of New York (SUNY) Downstate Medical Center Brooklyn, NY, USA
| | - Robert K S Wong
- Robert F. Furchgott Center for Neural and Behavioral Science and Department of Physiology and Pharmacology, State University of New York (SUNY) Downstate Medical Center Brooklyn, NY, USA
| | - Riccardo Bianchi
- Robert F. Furchgott Center for Neural and Behavioral Science and Department of Physiology and Pharmacology, State University of New York (SUNY) Downstate Medical Center Brooklyn, NY, USA
| | - Khalid Iqbal
- Department of Neurochemistry and SUNY Downstate/NYSIBR Center for Developmental Neuroscience, New York State Institute for Basic Research (NYSIBR) Staten Island, NY, USA
| |
Collapse
|
96
|
Abstract
PURPOSE OF REVIEW Suboptimal sleep has been reported as both a comorbidity and risk factor for the development of Alzheimer's disease. Previous research suggests that beta-amyloid (Aβ) may be central to this association, with reports indicating a bi-directional relationship between sleep and Aβ. Here, we review recent animal and human studies investigating the relationship between sleep and measures of Aβ, and explore the potential mechanisms underlying this association. RECENT FINDINGS Two recent animal studies have provided further support for a bi-directional relationship between sleep and Aβ. In addition, five recent human studies support the notion that higher brain Aβ is linked to poor sleep in cognitively healthy individuals. One of the recent human studies utilized polysomnography to link brain Aβ to a disruption in slow wave activity during sleep, which in turn was associated with decreased hippocampal-dependent memory. SUMMARY Recent findings indicate that poor sleep is a risk factor for brain Aβ deposition, and Aβ deposition contributes to sleep disruption. Through the conduct of more mechanistic studies, and both longitudinal and intervention human studies, we can further elucidate the clearly complex nature of the relationship between sleep and Aβ.
Collapse
|
97
|
Abstract
Authors: Kevin D Graber, MD, Jeffrey Buchhalter, MD, PhD, Elson So, MD, Rainer Surges, MD, Detlev Boison, PhD, Franck Kalume, PhD, Cyndi Wright, Brian Gehlbach, MD, Jeff Noebels, MD, PhD, Vicky Whittemore, PhD, Elizabeth J. Donner, MD, MSc, Tom Stanton, MPP, Henry Smithson, MD, Jane Hanna, Masud Seyal, MD, PhD, Philippe Ryvlin, MD, PhD The third biannual Partners Against Mortality in Epilepsy (PAME) conference was held in Alexandria, VA from June 23-26, 2016. This was an intimate meeting of clinical and basic scientists, clinicians, people affected by Sudden Unexpected Death in Epilepsy Patients (SUDEP) in a loved one, people living with epilepsy and patient advocate organizations. Plenary sessions have been summarized by moderators, including: 1) Mortality in people with epilepsy: epidemiology and surveillance. 2) Mortality in children. 3) What do we know about the factors that predispose certain people to die from a seizure? 4) What are the events that occur during and after a seizure that cause a death in SUDEP? 5) What are the options for prevention now and in the future? 6) Advocacy perspectives: how can we speed up awareness and prevention? 7) Updates and discussion on select programs in mortality research. Breakout sessions allowed for a more focused audience. Those summarized here are: Frequent non-SUDEP causes of mortality in people with epilepsy; Mechanisms of SUDEP; Lessons learned in grief and how to better support families; Future directions for research to impact prevention; and How do we improve SUDEP risk disclosure? While significant progress has been made with review of human mortality in epilepsy and study of animal models, this meeting emphasized the need for: better understandings of the epidemiology of SUDEP, advances in the understandings of mechanisms, continued search for biomarkers and preventative measures, patient education, increased awareness, continued advocacy for patient and family support and research funding.
Collapse
|