201
|
Exercise-Induced Cognitive Improvement Is Associated with Sodium Channel-Mediated Excitability in APP/PS1 Mice. Neural Plast 2020; 2020:9132720. [PMID: 32256560 PMCID: PMC7103997 DOI: 10.1155/2020/9132720] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/16/2020] [Accepted: 02/19/2020] [Indexed: 12/19/2022] Open
Abstract
Elevated brain activation, or hyperexcitability, induces cognitive impairment and confers an increased risk of Alzheimer's disease (AD). Blocking the overexcitation of the neural network may be a promising new strategy to prevent, halt, and even reverse this condition. Physical exercise has been shown to be an effective cognitive enhancer that reduces the risk of AD in elderly individuals, but the underlying mechanisms are far from being fully understood. We explored whether long-term treadmill exercise attenuates amyloid precursor protein (APP)/presenilin-1 (PS1) mutation-induced aberrant network activity and thus improves cognition by altering the numbers and/or distribution of voltage-gated sodium channels (Nav) in transgenic mice. APP/PS1 mice aged 2, 3.5, 5, 6.5, 8, and 9 months underwent treadmill exercise with different durations or at different stages of AD. The alterations in memory, electroencephalogram (EEG) recordings, and expression levels and distributions of Nav functional members (Nav1.1α, Nav1.2, Nav1.6, and Navβ2) were evaluated. The results revealed that treadmill exercise with 12- and 24-week durations 1) induced significant improvement in novel object recognition (NOR) memory and Morris water maze (MWM) spatial memory; 2) partially reduced abnormal spike activity; and 3) redressed the disturbed cellular distribution of Nav1.1α, aberrant Navβ2 cleavage augmentation, and Nav1.6 upregulation. Additionally, APP/PS1 mice in the 24-week exercise group showed better performance in the NOR task and a large decrease in Nav1.6 expression, which was close to the wild-type level. This study suggests that exercise improves cognition and neural activity by altering the numbers and distribution of hippocampal Nav in APP/PS1 mice. Long-term treadmill exercise, for about 24 weeks, starting in the preclinical stage, is a promising therapeutic strategy for preventing AD and halting its progress.
Collapse
|
202
|
Vyas Y, Montgomery JM, Cheyne JE. Hippocampal Deficits in Amyloid-β-Related Rodent Models of Alzheimer's Disease. Front Neurosci 2020; 14:266. [PMID: 32317913 PMCID: PMC7154147 DOI: 10.3389/fnins.2020.00266] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/09/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that is the most common cause of dementia. Symptoms of AD include memory loss, disorientation, mood and behavior changes, confusion, unfounded suspicions, and eventually, difficulty speaking, swallowing, and walking. These symptoms are caused by neuronal degeneration and cell loss that begins in the hippocampus, and later in disease progression spreading to the rest of the brain. While there are some medications that alleviate initial symptoms, there are currently no treatments that stop disease progression. Hippocampal deficits in amyloid-β-related rodent models of AD have revealed synaptic, behavioral and circuit-level defects. These changes in synaptic function, plasticity, neuronal excitability, brain connectivity, and excitation/inhibition imbalance all have profound effects on circuit function, which in turn could exacerbate disease progression. Despite, the wealth of studies on AD pathology we don't yet have a complete understanding of hippocampal deficits in AD. With the increasing development of in vivo recording techniques in awake and freely moving animals, future studies will extend our current knowledge of the mechanisms underpinning how hippocampal function is altered in AD, and aid in progression of treatment strategies that prevent and/or delay AD symptoms.
Collapse
Affiliation(s)
| | - Johanna M. Montgomery
- Department of Physiology, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Juliette E. Cheyne
- Department of Physiology, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| |
Collapse
|
203
|
Alshuaib S, Mosaddeghi J, Lin JW. Effects of levetiracetam on axon excitability and synaptic transmission at the crayfish neuromuscular junction. Synapse 2020; 74:e22154. [PMID: 32189403 DOI: 10.1002/syn.22154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/24/2020] [Accepted: 03/15/2020] [Indexed: 11/06/2022]
Abstract
Levetiracetam (LEV) is a widely prescribed antiepileptic drug, but its actions on neuronal function are not fully characterized. Since this drug is believed to enter neurons by binding to a vesicular protein during endocytosis, we used motor axons of the crayfish opener neuromuscular junction to examine potential impacts of LEV on axon excitability. Two electrode current clamp from the inhibitory axon of the opener showed that LEV reduced action potential (AP) amplitude (APamp ) and suppressed synaptic transmission, although the latter occurred with a longer delay than the reduction in APamp . Comparison of antidromic and orthodromic conducting APs in LEV suggested that this drug preferentially reduced excitability of the proximal axon, despite the expectation that it entered the axon at the terminals and should affect the distal branches first. Results presented here suggest that LEV modulates axonal excitability, which may in turn contribute to its antiepileptic effects.
Collapse
Affiliation(s)
| | | | - Jen-We Lin
- Department of Biology, Boston University, Boston, MA, USA
| |
Collapse
|
204
|
Abstract
Alzheimer's disease is a chronic neurodegenerative devastating disorder affecting a high percentage of the population over 65 years of age and causing a relevant emotional, social, and economic burden. Clinically, it is characterized by a prominent cognitive deficit associated with language and behavioral impairments. The molecular pathogenesis of Alzheimer's disease is multifaceted and involves changes in neurotransmitter levels together with alterations of inflammatory, oxidative, hormonal, and synaptic pathways, which may represent a drug target for both prevention and treatment; however, an effective treatment for Alzheimer's disease still represents an unmet goal. As neurotrophic factors participate in the modulation of the above-mentioned pathways, they have been highlighted as critical contributors of Alzheimer's disease etiology, whose modulation might be beneficial for Alzheimer's disease. We focused on the neurotrophin brain-derived neurotrophic factor, providing several lines of evidence pointing to brain-derived neurotrophic factor as a plausible endophenotype of cognitive deficits in Alzheimer's disease, illustrating some of the most recent possibilities to modulate the expression of this neurotrophin in the brain in an attempt to ameliorate cognition and delay the progression of Alzheimer's disease. This review shows that otherwise disparate pharmacologic or non-pharmacologic approaches converge on brain-derived neurotrophic factor, providing a means whereby apparently unrelated medical approaches may nevertheless produce similar synaptic and cognitive outcomes in Alzheimer's disease pathogenesis, suggesting that brain-derived neurotrophic factor-based synaptic repair may represent a modifying strategy to ameliorate cognition in Alzheimer's disease.
Collapse
|
205
|
Sen A, Jette N, Husain M, Sander JW. Epilepsy in older people. Lancet 2020; 395:735-748. [PMID: 32113502 DOI: 10.1016/s0140-6736(19)33064-8] [Citation(s) in RCA: 189] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/23/2019] [Accepted: 11/29/2019] [Indexed: 02/07/2023]
Abstract
Globally, as populations age there will be challenges and opportunities to deliver optimal health care to senior citizens. Epilepsy, a condition characterised by spontaneous recurrent seizures, is common in older adults (aged >65 years) and yet has received comparatively little attention in this age group. In this Review, we evaluate the underlying causes of epilepsy in older people, explore difficulties in establishing a diagnosis of epilepsy in this population, discuss appropriate antiseizure medications, and evaluate potential surgical treatment options. We consider cognitive, psychological, and psychosocial comorbidities and the effect that epilepsy might have on an older person's broader social or care network in high-income versus middle-income and low-income countries. We emphasise the need for clinical trials to be more inclusive of older people with epilepsy to help inform therapeutic decision making and discuss whether measures to improve vascular risk factors might be an important strategy to reduce the probability of developing epilepsy.
Collapse
Affiliation(s)
- Arjune Sen
- Oxford Epilepsy Research Group, National Institute for Health Research Oxford Biomedical Research Centre, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, UK.
| | - Nathalie Jette
- Departments of Neurology and Population Health Sciences & Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Masud Husain
- Department of Psychology, University of Oxford, Oxford UK
| | - Josemir W Sander
- National Institute for Health Research, Biomedical Research Centre, University College London Hospitals, UCL Queen Square Institute of Neurology, London, UK; Chalfont Centre for Epilepsy, Chalfont St Peter, UK; Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, Netherlands
| |
Collapse
|
206
|
Cai Z, Li S, Zhang W, Pracitto R, Wu X, Baum E, Finnema SJ, Holden D, Toyonaga T, Lin SF, Lindemann M, Shirali A, Labaree DC, Ropchan J, Nabulsi N, Carson RE, Huang Y. Synthesis and Preclinical Evaluation of an 18F-Labeled Synaptic Vesicle Glycoprotein 2A PET Imaging Probe: [ 18F]SynVesT-2. ACS Chem Neurosci 2020; 11:592-603. [PMID: 31961649 DOI: 10.1021/acschemneuro.9b00618] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Synaptic vesicle glycoprotein 2A (SV2A) is a 12-pass transmembrane glycoprotein ubiquitously expressed in presynaptic vesicles. In vivo imaging of SV2A using PET has potential applications in the diagnosis and prognosis of a variety of neuropsychiatric diseases, e.g., Alzheimer's disease, Parkinson's disease, schizophrenia, multiple sclerosis, autism, epilepsy, stroke, traumatic brain injury, post-traumatic stress disorder, depression, etc. Herein, we report the synthesis and evaluation of a new 18F-labeled SV2A PET imaging probe, [18F]SynVesT-2, which possesses fast in vivo binding kinetics and high specific binding signals in non-human primate brain.
Collapse
Affiliation(s)
- Zhengxin Cai
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Songye Li
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Wenjie Zhang
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Richard Pracitto
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Xiaoai Wu
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Evan Baum
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Sjoerd J. Finnema
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Daniel Holden
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Takuya Toyonaga
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Shu-fei Lin
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Marcel Lindemann
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Anupama Shirali
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - David C. Labaree
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Jim Ropchan
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Nabeel Nabulsi
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Richard E. Carson
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| |
Collapse
|
207
|
Calvo-Flores Guzmán B, Kim S, Chawdhary B, Peppercorn K, Tate WP, Waldvogel HJ, Faull RLM, Montgomery J, Kwakowsky A. Amyloid-Beta 1-42 -Induced Increase in GABAergic Tonic Conductance in Mouse Hippocampal CA1 Pyramidal Cells. Molecules 2020; 25:molecules25030693. [PMID: 32041202 PMCID: PMC7037727 DOI: 10.3390/molecules25030693] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/03/2020] [Accepted: 02/03/2020] [Indexed: 01/23/2023] Open
Abstract
Alzheimer’s disease (AD) is a complex and chronic neurodegenerative disorder that involves a progressive and severe decline in cognition and memory. During the last few decades a considerable amount of research has been done in order to better understand tau-pathology, inflammatory activity and neuronal synapse loss in AD, all of them contributing to cognitive decline. Early hippocampal network dysfunction is one of the main factors associated with cognitive decline in AD. Much has been published about amyloid-beta1-42 (Aβ1-42)-mediated excitotoxicity in AD. However, increasing evidence demonstrates that the remodeling of the inhibitory gamma-aminobutyric acid (GABAergic) system contributes to the excitatory/inhibitory (E/I) disruption in the AD hippocampus, but the underlying mechanisms are not well understood. In the present study, we show that hippocampal injection of Aβ1-42 is sufficient to induce cognitive deficits 7 days post-injection. We demonstrate using in vitro whole-cell patch-clamping an increased inhibitory GABAergic tonic conductance mediated by extrasynaptic type A GABA receptors (GABAARs), recorded in the CA1 region of the mouse hippocampus following Aβ1-42 micro injection. Such alterations in GABA neurotransmission and/or inhibitory GABAARs could have a significant impact on both hippocampal structure and function, causing E/I balance disruption and potentially contributing to cognitive deficits in AD.
Collapse
Affiliation(s)
- Beatriz Calvo-Flores Guzmán
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (B.C.-F.G.); (S.K.); (B.C.); (H.J.W.); (R.L.F.)
| | - SooHyun Kim
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (B.C.-F.G.); (S.K.); (B.C.); (H.J.W.); (R.L.F.)
| | - Bhavya Chawdhary
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (B.C.-F.G.); (S.K.); (B.C.); (H.J.W.); (R.L.F.)
| | - Katie Peppercorn
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand; (K.P.); (W.P.T.)
| | - Warren P Tate
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand; (K.P.); (W.P.T.)
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (B.C.-F.G.); (S.K.); (B.C.); (H.J.W.); (R.L.F.)
| | - Richard LM Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (B.C.-F.G.); (S.K.); (B.C.); (H.J.W.); (R.L.F.)
| | - Johanna Montgomery
- Centre for Brain Research, Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand;
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (B.C.-F.G.); (S.K.); (B.C.); (H.J.W.); (R.L.F.)
- Correspondence: ; Tel.: +64-9923-9346
| |
Collapse
|
208
|
Sánchez-Rodríguez I, Djebari S, Temprano-Carazo S, Vega-Avelaira D, Jiménez-Herrera R, Iborra-Lázaro G, Yajeya J, Jiménez-Díaz L, Navarro-López JD. Hippocampal long-term synaptic depression and memory deficits induced in early amyloidopathy are prevented by enhancing G-protein-gated inwardly rectifying potassium channel activity. J Neurochem 2020; 153:362-376. [PMID: 31875959 PMCID: PMC7217154 DOI: 10.1111/jnc.14946] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 12/06/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022]
Abstract
Hippocampal synaptic plasticity disruption by amyloid‐β (Aβ) peptides + thought to be responsible for learning and memory impairments in Alzheimer's disease (AD) early stage. Failures in neuronal excitability maintenance seems to be an underlying mechanism. G‐protein‐gated inwardly rectifying potassium (GirK) channels control neural excitability by hyperpolarization in response to many G‐protein‐coupled receptors activation. Here, in early in vitro and in vivo amyloidosis mouse models, we study whether GirK channels take part of the hippocampal synaptic plasticity impairments generated by Aβ1–42. In vitro electrophysiological recordings from slices showed that Aβ1–42 alters synaptic plasticity by switching high‐frequency stimulation (HFS) induced long‐term potentiation (LTP) to long‐term depression (LTD), which led to in vivo hippocampal‐dependent memory deficits. Remarkably, selective pharmacological activation of GirK channels with ML297 rescued both HFS‐induced LTP and habituation memory from Aβ1–42 action. Moreover, when GirK channels were specifically blocked by Tertiapin‐Q, their activation with ML297 failed to rescue LTP from the HFS‐dependent LTD induced by Aβ1–42. On the other hand, the molecular analysis of the recorded slices by western blot showed that the expression of GIRK1/2 subunits, which form the prototypical GirK channel in the hippocampus, was not significantly regulated by Aβ1–42. However, immunohistochemical examination of our in vivo amyloidosis model showed Aβ1–42 to down‐regulate hippocampal GIRK1 subunit expression. Together, our results describe an Aβ‐mediated deleterious synaptic mechanism that modifies the induction threshold for hippocampal LTP/LTD and underlies memory alterations observed in amyloidosis models. In this scenario, GirK activation assures memory formation by preventing the transformation of HFS‐induced LTP into LTD. ![]()
Collapse
Affiliation(s)
- Irene Sánchez-Rodríguez
- NeuroPhysiology & Behavior Laboratory, Centro Regional de Investigaciones Biomédicas, School of Medicine of Ciudad Real, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Souhail Djebari
- NeuroPhysiology & Behavior Laboratory, Centro Regional de Investigaciones Biomédicas, School of Medicine of Ciudad Real, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Sara Temprano-Carazo
- NeuroPhysiology & Behavior Laboratory, Centro Regional de Investigaciones Biomédicas, School of Medicine of Ciudad Real, University of Castilla-La Mancha, Ciudad Real, Spain
| | - David Vega-Avelaira
- Departamento de Ciencias Biomédicas Básicas, European University of Madrid, Madrid, Spain
| | - Raquel Jiménez-Herrera
- NeuroPhysiology & Behavior Laboratory, Centro Regional de Investigaciones Biomédicas, School of Medicine of Ciudad Real, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Guillermo Iborra-Lázaro
- NeuroPhysiology & Behavior Laboratory, Centro Regional de Investigaciones Biomédicas, School of Medicine of Ciudad Real, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Javier Yajeya
- Instituto de Neurociencias de Castilla y León, University of Salamanca, Salamanca, Spain
| | - Lydia Jiménez-Díaz
- NeuroPhysiology & Behavior Laboratory, Centro Regional de Investigaciones Biomédicas, School of Medicine of Ciudad Real, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Juan D Navarro-López
- NeuroPhysiology & Behavior Laboratory, Centro Regional de Investigaciones Biomédicas, School of Medicine of Ciudad Real, University of Castilla-La Mancha, Ciudad Real, Spain
| |
Collapse
|
209
|
Elia LP, Reisine T, Alijagic A, Finkbeiner S. Approaches to develop therapeutics to treat frontotemporal dementia. Neuropharmacology 2020; 166:107948. [PMID: 31962288 DOI: 10.1016/j.neuropharm.2020.107948] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/16/2019] [Accepted: 01/07/2020] [Indexed: 12/13/2022]
Abstract
Frontotemporal degeneration (FTD) is a complex disease presenting as a spectrum of clinical disorders with progressive degeneration of frontal and temporal brain cortices and extensive neuroinflammation that result in personality and behavior changes, and eventually, death. There are currently no effective therapies for FTD. While 60-70% of FTD patients are sporadic cases, the other 30-40% are heritable (familial) cases linked to mutations in several known genes. We focus here on FTD caused by mutations in the GRN gene, which encodes a secreted protein, progranulin (PGRN), that has diverse roles in regulating cell survival, immune responses, and autophagy and lysosome function in the brain. FTD-linked mutations in GRN reduce brain PGRN levels that lead to autophagy and lysosome dysfunction, TDP43 accumulation, excessive microglial activation, astrogliosis, and neuron death through still poorly understood mechanisms. PGRN insufficiency has also been linked to Alzheimer's disease (AD), and so the development of therapeutics for GRN-linked FTD that restore PGRN levels and function may have broader application for other neurodegenerative diseases. This review focuses on a strategy to increase PGRN to functional, healthy levels in the brain by identifying novel genetic and chemical modulators of neuronal PGRN levels. This article is part of the special issue entitled 'The Quest for Disease-Modifying Therapies for Neurodegenerative Disorders'.
Collapse
Affiliation(s)
- Lisa P Elia
- Center for Systems and Therapeutics and Taube/Koret Center for Neurodegenerative Disease Research, San Francisco, CA, USA; The J. David Gladstone Institutes, San Francisco, CA, USA.
| | - Terry Reisine
- Independent Scientific Consultant, Santa Cruz, CA, USA
| | - Amela Alijagic
- Center for Systems and Therapeutics and Taube/Koret Center for Neurodegenerative Disease Research, San Francisco, CA, USA; The J. David Gladstone Institutes, San Francisco, CA, USA
| | - Steven Finkbeiner
- Center for Systems and Therapeutics and Taube/Koret Center for Neurodegenerative Disease Research, San Francisco, CA, USA; The J. David Gladstone Institutes, San Francisco, CA, USA; Departments of Neurology and Physiology, UCSF, San Francisco, CA, USA.
| |
Collapse
|
210
|
Peña-Ortega F. Brain Arrhythmias Induced by Amyloid Beta and Inflammation: Involvement in Alzheimer’s Disease and Other Inflammation-related Pathologies. Curr Alzheimer Res 2020; 16:1108-1131. [DOI: 10.2174/1567205017666191213162233] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 12/13/2022]
Abstract
A variety of neurological diseases, including Alzheimer’s disease (AD), involve amyloid beta (Aβ) accumulation and/or neuroinflammation, which can alter synaptic and neural circuit functions. Consequently, these pathological conditions induce changes in neural network rhythmic activity (brain arrhythmias), which affects many brain functions. Neural network rhythms are involved in information processing, storage and retrieval, which are essential for memory consolidation, executive functioning and sensory processing. Therefore, brain arrhythmias could have catastrophic effects on circuit function, underlying the symptoms of various neurological diseases. Moreover, brain arrhythmias can serve as biomarkers for a variety of brain diseases. The aim of this review is to provide evidence linking Aβ and inflammation to neural network dysfunction, focusing on alterations in brain rhythms and their impact on cognition and sensory processing. I reviewed the most common brain arrhythmias characterized in AD, in AD transgenic models and those induced by Aβ. In addition, I reviewed the modulations of brain rhythms in neuroinflammatory diseases and those induced by immunogens, interleukins and microglia. This review reveals that Aβ and inflammation produce a complex set of effects on neural network function, which are related to the induction of brain arrhythmias and hyperexcitability, both closely related to behavioral alterations. Understanding these brain arrhythmias can help to develop therapeutic strategies to halt or prevent these neural network alterations and treat not only the arrhythmias but also the symptoms of AD and other inflammation-related pathologies.
Collapse
Affiliation(s)
- Fernando Peña-Ortega
- Departamento de Neurobiologia del Desarrollo y Neurofisiologia, Instituto de Neurobiologia, Universidad Nacional Autonoma de Mexico, Queretaro, Qro., 76230, Mexico
| |
Collapse
|
211
|
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: 3.6] [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
|
212
|
Early restoration of parvalbumin interneuron activity prevents memory loss and network hyperexcitability in a mouse model of Alzheimer's disease. Mol Psychiatry 2020; 25:3380-3398. [PMID: 31431685 PMCID: PMC7714697 DOI: 10.1038/s41380-019-0483-4] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 05/09/2019] [Accepted: 06/20/2019] [Indexed: 02/06/2023]
Abstract
Neuronal network dysfunction is increasingly recognized as an early symptom in Alzheimer's disease (AD) and may provide new entry points for diagnosis and intervention. Here, we show that amyloid-beta-induced hyperexcitability of hippocampal inhibitory parvalbumin (PV) interneurons importantly contributes to neuronal network dysfunction and memory impairment in APP/PS1 mice, a mouse model of increased amyloidosis. We demonstrate that hippocampal PV interneurons become hyperexcitable at ~16 weeks of age, when no changes are observed yet in the intrinsic properties of pyramidal cells. This hyperexcitable state of PV interneurons coincides with increased inhibitory transmission onto hippocampal pyramidal neurons and deficits in spatial learning and memory. We show that treatment aimed at preventing PV interneurons from becoming hyperexcitable is sufficient to restore PV interneuron properties to wild-type levels, reduce inhibitory input onto pyramidal cells, and rescue memory deficits in APP/PS1 mice. Importantly, we demonstrate that early intervention aimed at restoring PV interneuron activity has long-term beneficial effects on memory and hippocampal network activity, and reduces amyloid plaque deposition, a hallmark of AD pathology. Taken together, these findings suggest that early treatment of PV interneuron hyperactivity might be clinically relevant in preventing memory decline and delaying AD progression.
Collapse
|
213
|
Chen Y, Gao X, Liu Q, Zeng L, Zhang K, Mu K, Zhang D, Zou H, Wu N, Ou J, Wang Q, Mao S. Alpha-asarone improves cognitive function of aged rats by alleviating neuronal excitotoxicity via GABAA receptors. Neuropharmacology 2020; 162:107843. [DOI: 10.1016/j.neuropharm.2019.107843] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/03/2019] [Accepted: 11/04/2019] [Indexed: 12/20/2022]
|
214
|
What electrophysiology tells us about Alzheimer's disease: a window into the synchronization and connectivity of brain neurons. Neurobiol Aging 2020; 85:58-73. [DOI: 10.1016/j.neurobiolaging.2019.09.008] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/27/2019] [Accepted: 09/14/2019] [Indexed: 01/14/2023]
|
215
|
Leparulo A, Mahmud M, Scremin E, Pozzan T, Vassanelli S, Fasolato C. Dampened Slow Oscillation Connectivity Anticipates Amyloid Deposition in the PS2APP Mouse Model of Alzheimer's Disease. Cells 2019; 9:cells9010054. [PMID: 31878336 PMCID: PMC7016892 DOI: 10.3390/cells9010054] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/17/2019] [Accepted: 12/20/2019] [Indexed: 12/21/2022] Open
Abstract
To fight Alzheimer's disease (AD), we should know when, where, and how brain network dysfunctions initiate. In AD mouse models, relevant information can be derived from brain electrical activity. With a multi-site linear probe, we recorded local field potentials simultaneously at the posterior-parietal cortex and hippocampus of wild-type and double transgenic AD mice, under anesthesia. We focused on PS2APP (B6.152H) mice carrying both presenilin-2 (PS2) and amyloid precursor protein (APP) mutations, at three and six months of age, before and after plaque deposition respectively. To highlight defects linked to either the PS2 or APP mutation, we included in the analysis age-matched PS2.30H and APP-Swedish mice, carrying each of the mutations individually. Our study also included PSEN2-/- mice. At three months, only predeposition B6.152H mice show a reduction in the functional connectivity of slow oscillations (SO) and in the power ratio between SO and delta waves. At six months, plaque-seeding B6.152H mice undergo a worsening of the low/high frequency power imbalance and show a massive loss of cortico-hippocampal phase-amplitude coupling (PAC) between SO and higher frequencies, a feature shared with amyloid-free PS2.30H mice. We conclude that the PS2 mutation is sufficient to impair SO PAC and accelerate network dysfunctions in amyloid-accumulating mice.
Collapse
Affiliation(s)
- Alessandro Leparulo
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (A.L.); (M.M.); (E.S.); (T.P.)
| | - Mufti Mahmud
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (A.L.); (M.M.); (E.S.); (T.P.)
| | - Elena Scremin
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (A.L.); (M.M.); (E.S.); (T.P.)
| | - Tullio Pozzan
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (A.L.); (M.M.); (E.S.); (T.P.)
- Neuroscience Institute-Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
- Venetian Institute of Molecular Medicine (VIMM), Via G. Orus 2B, 35129 Padua, Italy
| | - Stefano Vassanelli
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (A.L.); (M.M.); (E.S.); (T.P.)
- Padua Neuroscience Center (PNC), University of Padua, Via G. Orus 2B, 35129 Padua, Italy
- Correspondence: (S.V.); (C.F.); Tel.: +39-049-8275337 (S.V.); +39-049-8276065 (C.F.); Fax: +39-049-8276049 (S.V.); +39-049-8276049 (C.F.)
| | - Cristina Fasolato
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (A.L.); (M.M.); (E.S.); (T.P.)
- Correspondence: (S.V.); (C.F.); Tel.: +39-049-8275337 (S.V.); +39-049-8276065 (C.F.); Fax: +39-049-8276049 (S.V.); +39-049-8276049 (C.F.)
| |
Collapse
|
216
|
Beckman M, Knox K, Koneval Z, Smith C, Jayadev S, Barker-Haliski M. Loss of presenilin 2 age-dependently alters susceptibility to acute seizures and kindling acquisition. Neurobiol Dis 2019; 136:104719. [PMID: 31862541 DOI: 10.1016/j.nbd.2019.104719] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/17/2019] [Accepted: 12/16/2019] [Indexed: 01/15/2023] Open
Abstract
Patients with Alzheimer's disease (AD) experience seizures at higher rates than the general population of that age, suggesting an underexplored role of hyperexcitability in AD. Genetic variants in presenilin (PSEN) 1 and 2 genes lead to autosomal dominant early-onset AD (ADAD); patients with PSEN gene variants also report seizures. Pharmacological control of seizures in AD may be disease-modifying. Preclinical efficacy of FDA-approved antiseizure drugs (ASDs) is well defined in young adult rodents; however, the efficacy of ASDs in aged rodents with chronic seizures is less clear. The mechanism by which ADAD genes lead to AD remains unclear, and even less studied is the pathogenesis of epilepsy in AD. PSEN variants generally all result in a biochemical loss of function (De Strooper, 2007). We herein determined whether well-established models of acute and chronic seizure could be used to explore the relationship between AD genes and seizures through investigating whether loss of normal PSEN2 function age-dependently influenced susceptibility to seizures and/or corneal kindling acquisition. PSEN2 knockout (KO) and age-matched wild-type (WT) mice were screened from 2- to 10-months-old to establish age-dependent focal seizure threshold. Additionally, PSEN2 KO and WT mice aged 2- and 8-months-old underwent corneal kindling such that mice were aged 3- and 9-months old at the beginning of ASD efficacy testing. We then defined the dose-dependent efficacy of mechanistically distinct ASDs on kindled seizures of young versus aged mice to better understand the applicability of corneal kindling to real-world use for geriatric patients. PSEN2 KO mice demonstrated early-life reductions in seizure threshold. However, kindling acquisition was delayed in 2-month-old PSEN2 KO versus WT mice. Young male WT mice took 24.3 ± 1.3 (S.E.M.) stimulations to achieve kindling criterion, whereas age-matched PSEN2 KO male mice took 41.2 ± 1.1 stimulations (p < .0001). The rate of kindling acquisition of 8-month-old mice was no longer different from WT. This study demonstrates that loss of normal PSEN2 function is associated with age-dependent changes in the in vivo susceptibility to acute seizures and kindling. Loss of normal PSEN2 function may be an underexplored molecular contributor to seizures. The use of validated models of chronic seizures in aged rodents may uncover age-related changes in susceptibility to epileptogenesis and/or ASD efficacy in mice with AD-associated genotypes, which may benefit the management of seizures in AD.
Collapse
Affiliation(s)
- Megan Beckman
- Department of Pharmacy, School of Pharmacy, University of Washington, United States of America
| | - Kevin Knox
- Department of Pharmacy, School of Pharmacy, University of Washington, United States of America
| | - Zachery Koneval
- Department of Pharmacy, School of Pharmacy, University of Washington, United States of America
| | - Carole Smith
- Department of Neurology, School of Medicine, University of Washington, United States of America
| | - Suman Jayadev
- Department of Neurology, School of Medicine, University of Washington, United States of America
| | - Melissa Barker-Haliski
- Department of Pharmacy, School of Pharmacy, University of Washington, United States of America.
| |
Collapse
|
217
|
Baker J, Libretto T, Henley W, Zeman A. A Longitudinal Study of Epileptic Seizures in Alzheimer's Disease. Front Neurol 2019; 10:1266. [PMID: 31866927 PMCID: PMC6904279 DOI: 10.3389/fneur.2019.01266] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 11/15/2019] [Indexed: 12/19/2022] Open
Abstract
The prevalence of epileptic seizures is increased in patients in the clinical stages of Alzheimer's disease (AD) when compared to age-matched cognitively normal populations. In previously reported work from the Presentation of Epileptic Seizures in Dementia (PrESIDe) study, we identified a clinical suspicion of epilepsy in between 12.75 and 28.43% of patients with AD recruited from a memory clinic. EEGs were not performed in this study. Patients with epilepsy performed similarly to patients without epilepsy on cognitive testing at the time of recruitment but were more impaired on two measures of everyday functioning [Cambridge Behavioral Inventory—Revised and Clinical Dementia Rating (CBI-R and CDR)]. On repeated testing in this 12-month follow-up study, patients in whom a suspicion of epilepsy was identified performed significantly worse on cognitive function testing (p = 0.028) in addition to maintaining a difference on the informant questionnaires (CBI-R p < 0.001, CDR p = 0.020). These findings suggest that seizures in this population could be a marker of a more rapid decline and worse prognosis.
Collapse
Affiliation(s)
- John Baker
- Cognitive and Behavioral Neurology, College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Tina Libretto
- NIHR Exeter Clinical Research Facility, Royal Devon and Exeter NHS Foundation Trust, Exeter, United Kingdom
| | - William Henley
- College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Adam Zeman
- Cognitive and Behavioral Neurology, College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
218
|
Wang S, Wang ZI, Tang Y, Alexopoulos AV, Chen C, Katagiri M, Aung T, Najm IM, Ding M, Wang S, Chauvel P. Localization value of subclinical seizures on scalp video‐EEG in epilepsy presurgical evaluation. Epilepsia 2019; 60:2477-2485. [PMID: 31755095 DOI: 10.1111/epi.16383] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 10/18/2019] [Accepted: 10/18/2019] [Indexed: 02/05/2023]
Affiliation(s)
- Shan Wang
- Department of Neurology Epilepsy Center Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
- Epilepsy Center Neurological Institute Cleveland Clinic Cleveland OH USA
| | - Z. Irene Wang
- Epilepsy Center Neurological Institute Cleveland Clinic Cleveland OH USA
| | - Yingying Tang
- Epilepsy Center Neurological Institute Cleveland Clinic Cleveland OH USA
- Department of Neurology West China Hospital Sichuan University Chengdu China
| | | | - Cong Chen
- Department of Neurology Epilepsy Center Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
| | - Masaya Katagiri
- Epilepsy Center Neurological Institute Cleveland Clinic Cleveland OH USA
| | - Thandar Aung
- Epilepsy Center Neurological Institute Cleveland Clinic Cleveland OH USA
| | - Imad M. Najm
- Epilepsy Center Neurological Institute Cleveland Clinic Cleveland OH USA
| | - Meiping Ding
- Department of Neurology Epilepsy Center Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
| | - Shuang Wang
- Department of Neurology Epilepsy Center Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
| | - Patrick Chauvel
- Epilepsy Center Neurological Institute Cleveland Clinic Cleveland OH USA
| |
Collapse
|
219
|
Rozycka A, Charzynska A, Misiewicz Z, Maciej Stepniewski T, Sobolewska A, Kossut M, Liguz-Lecznar M. Glutamate, GABA, and Presynaptic Markers Involved in Neurotransmission Are Differently Affected by Age in Distinct Mouse Brain Regions. ACS Chem Neurosci 2019; 10:4449-4461. [PMID: 31556991 DOI: 10.1021/acschemneuro.9b00220] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Molecular synaptic aging perturbs neurotransmission and decreases the potential for neuroplasticity. The direction and degree of changes observed in aging are often region or cell specific, hampering the generalization of age-related effects. Using real-time PCR and Western blot analyses, we investigated age-related changes in several presynaptic markers (Vglut1, Vglut2, Gad65, Gad67, Vgat, synaptophysin) involved in the initial steps of glutamatergic and GABAergic neurotransmission, in several cortical regions, in young (3-4 months old), middle-aged (1 year old), and old (2 years old) mice. We found age-related changes mainly in protein levels while, apart from the occipital cortex, virtually no significant changes in mRNA levels were detected, which suggests that aging acts on the investigated markers mainly through post-transcriptional mechanisms depending on the brain region. Principal component analysis (PCA) of protein data revealed that each brain region possessed a type of "biochemical distinctiveness" (each analyzed brain region was best characterized by higher variability level of a particular synaptic marker) that was lost with age. Analysis of glutamate and γ-aminobutyric acid (GABA) levels in aging suggested that mechanisms keeping an overall balance between the two amino acids in the brain are weakened in the hippocampus. Our results unravel the differences in mRNA/protein interactions in the aging brain.
Collapse
Affiliation(s)
- Aleksandra Rozycka
- Laboratory of Neuroplasticity, Nencki Institute of Experimental Biology Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland
| | - Agata Charzynska
- Laboratory of Bioinformatics, Neurobiology Center, Nencki Institute of Experimental Biology Polish Academy of Sciences, 3 Pasteur Str., 02-093 Warsaw, Poland
| | - Zuzanna Misiewicz
- Laboratory of Neuroplasticity, Nencki Institute of Experimental Biology Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki FI-00014, Finland
| | - Tomasz Maciej Stepniewski
- Laboratory of Neuroplasticity, Nencki Institute of Experimental Biology Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland
- Research Programme on Biomedical Informatics (GRIB) - Department of Experimental and Health Sciences, Hospital del Mar Medical Research Institute, Pompeu Fabra University, 08002 Barcelona, Spain
| | - Alicja Sobolewska
- Department of Neurochemistry, Institute of Psychiatry and Neurology, 9 Sobieskiego Str., 02-957 Warsaw, Poland
| | - Malgorzata Kossut
- Laboratory of Neuroplasticity, Nencki Institute of Experimental Biology Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland
- Faculty of Psychology, SWPS University of Social Sciences and Humanities, 03-815 Warsaw, Poland
| | - Monika Liguz-Lecznar
- Laboratory of Neuroplasticity, Nencki Institute of Experimental Biology Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland
| |
Collapse
|
220
|
Fessel J. Prevention of Alzheimer's disease by treating mild cognitive impairment with combinations chosen from eight available drugs. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2019; 5:780-788. [PMID: 31763432 PMCID: PMC6861553 DOI: 10.1016/j.trci.2019.09.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Several hundred clinical trials of initially promising drugs have failed to produce meaningful clinical improvement of Alzheimer's disease (AD), which is probably because there are at least 25 biochemical pathways known to be aberrant that underpin the disease, and unless there is a single drug that addresses all or most of them, even promising drugs if given alone are unlikely to succeed. Because so many pathways are potentially at fault, it is quite possible that no treatment might succeed. However, because amnestic mild cognitive impairment (aMCI) often precedes AD and, assuming that those with aMCI who progress to AD commence with insufficient risk factors for AD but accrue them later, then it is likely that fewer pathways need addressing in aMCI than in AD to either prevent progression of aMCI to AD or effect its reversion. Published reports show that eight drugs, that is, dantrolene, erythropoietin, lithium, memantine, minocycline, piracetam, riluzole, and silymarin, address many of the pathways underlying MCI and AD. Among those eight drugs, combinations between either two or three of them have combined nonoverlapping actions that benefit enough of the approximately 25 pathways at fault so that their convergent efficacy has the potential to prevent aMCI from progressing to AD. The combinations should be subjected to a clinical trial in persons with aMCI to establish their safety and efficacy.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Department of Medicine, University of California San Francisco, San Francisco, CA
| |
Collapse
|
221
|
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: 6.8] [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
|
222
|
Synthesis, in-vitro cholinesterase inhibition, in-vivo anticonvulsant activity and in-silico exploration of N-(4-methylpyridin-2-yl)thiophene-2-carboxamide analogs. Bioorg Chem 2019; 92:103216. [DOI: 10.1016/j.bioorg.2019.103216] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 11/17/2022]
|
223
|
Werner CT, Williams CJ, Fermelia MR, Lin DT, Li Y. Circuit Mechanisms of Neurodegenerative Diseases: A New Frontier With Miniature Fluorescence Microscopy. Front Neurosci 2019; 13:1174. [PMID: 31736701 PMCID: PMC6834692 DOI: 10.3389/fnins.2019.01174] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 10/17/2019] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases (NDDs), such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS), and frontotemporal dementia (FTD), are devastating age-associated brain disorders. Significant efforts have been made to uncover the molecular and cellular pathogenic mechanisms that underlie NDDs. However, our understanding of the neural circuit mechanisms that mediate NDDs and associated symptomatic features have been hindered by technological limitations. Our inability to identify and track individual neurons longitudinally in subcortical brain regions that are preferentially targeted in NDDs has left gaping holes in our knowledge of NDDs. Recent development and advancement of the miniature fluorescence microscope (miniscope) has opened up new avenues for examining spatially and temporally coordinated activity from hundreds of cells in deep brain structures in freely moving rodents. In the present mini-review, we examine the capabilities of current and future miniscope tools and discuss the innovative applications of miniscope imaging techniques that can push the boundaries of our understanding of neural circuit mechanisms of NDDs into new territories.
Collapse
Affiliation(s)
- Craig T Werner
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | | | - Mercedes R Fermelia
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, United States
| | - Da-Ting Lin
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States.,The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Yun Li
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, United States
| |
Collapse
|
224
|
Iron Overload Impairs Autophagy: Effects of Rapamycin in Ameliorating Iron-Related Memory Deficits. Mol Neurobiol 2019; 57:1044-1054. [PMID: 31664701 DOI: 10.1007/s12035-019-01794-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 09/22/2019] [Indexed: 12/15/2022]
Abstract
Over the years, iron accumulation in specific brain regions has been observed in normal aging and related to the pathogenesis of neurodegenerative disorders. Many neurodegenerative diseases may involve cognitive dysfunction, and we have previously shown that neonatal iron overload induces permanent cognitive deficits in adult rats and exacerbates age-associated memory decline. Autophagy is a catabolic pathway involved in the removal of toxic protein aggregates, which are a hallmark of neurodegenerative events. In the present study, we investigated whether iron accumulation would interfere with autophagy and also sought to determine the effects of rapamycin-induced stimulation of autophagy in attenuating iron-related cognitive deficits. Male Wistar rats received a single daily oral dose of vehicle or iron carbonyl (30 mg/kg) at postnatal days 12-14. In adulthood, they received daily intraperitoneal injections of vehicle or rapamycin (0.25 mg/kg) for 14 days. Results showed that iron given in the neonatal period impaired inhibitory avoidance memory and induced a decrease in proteins critically involved in the autophagy pathway, Beclin-1 and LC3, in the hippocampus. Rapamycin in the adulthood reversed iron-induced memory deficits, decreased the ratio phospho-mTOR/total mTOR, and recovered LC3 II levels in iron-treated rats. Our results suggest that iron accumulation, as observed in neurodegenerative disorders, hinders autophagy, which might play a role in iron-induced neurotoxicity. Rapamycin, by inducing authophagy, was able to ameliorate iron-induced cognitive impairments. These findings support the use of rapamycin as a potential neuroprotective treatment against the cognitive decline associated to neurodegenerative disorders.
Collapse
|
225
|
Ahn JH, Shin BN, Park JH, Lee TK, Park YE, Lee JC, Yang GE, Shin MC, Cho JH, Lee KC, Won MH, Kim H. Pre- and Post-Treatment with Novel Antiepileptic Drug Oxcarbazepine Exerts Neuroprotective Effect in the Hippocampus in a Gerbil Model of Transient Global Cerebral Ischemia. Brain Sci 2019; 9:brainsci9100279. [PMID: 31627311 PMCID: PMC6826395 DOI: 10.3390/brainsci9100279] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/11/2019] [Accepted: 10/16/2019] [Indexed: 01/01/2023] Open
Abstract
Oxcarbazepine, an antiepileptic drug, has been reported to modulate voltage-dependent sodium channels, and it is commonly used in epilepsy treatment. In this study, we investigated the neuroprotective effect of oxcarbazepine in the hippocampus after transient ischemia in gerbils. Gerbils randomly received oxcarbazepine 100 or 200 mg/kg before and after transient ischemia. We examined its neuroprotective effect in the cornu ammonis 1 subfield of the gerbil hippocampus at 5 days after transient ischemia by using cresyl violet staining, neuronal nuclei immunohistochemistry and Fluoro-Jade B histofluorescence staining for neuroprotection, and by using glial fibrillary protein and ionized calcium-binding adapter molecule 1 immunohistochemistry for reaction of astrocytes and microglia, respectively. Pre- and post-treatment with 200 mg/kg of oxcarbazepine, but not 100 mg/kg of oxcarbazepine, protected pyramidal neurons of the cornu ammonis 1 subfield from transient ischemic damage. In addition, pre- and post-treatment with oxcarbazepine (200 mg/kg) significantly ameliorated astrocytes and microglia activation in the ischemic cornu ammonis 1 subfield. In brief, our current results indicate that post-treatment as well as pre-treatment with 200 mg/kg of oxcarbazepine can protect neurons from ischemic insults via attenuation of the glia reaction.
Collapse
Affiliation(s)
- Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Korea.
| | - Bich Na Shin
- Department of Physiology, School of Medicine, Hallym University, Chuncheon, Gangwon 24252, Korea.
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju, Gyeongbuk 38066, Korea.
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Korea.
| | - Young Eun Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Korea.
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Korea.
| | - Go Eun Yang
- Department of Radiology, Kangwon National University Hospital, Chuncheon, Gangwon 24289, Korea.
| | - Myoung Cheol Shin
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Korea.
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Korea.
| | - Kyu Chang Lee
- Department of Anesthesiology and Pain Medicine, Chungju Hospital, Konkuk University School of Medicine, Chungju, Chungbuk 27376, Korea.
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Korea.
| | - Hyeyoung Kim
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Korea.
- Department of Anesthesiology and Pain Medicine, Chungju Hospital, Konkuk University School of Medicine, Chungju, Chungbuk 27376, Korea.
| |
Collapse
|
226
|
Singh B, Covelo A, Martell-Martínez H, Nanclares C, Sherman MA, Okematti E, Meints J, Teravskis PJ, Gallardo C, Savonenko AV, Benneyworth MA, Lesné SE, Liao D, Araque A, Lee MK. Tau is required for progressive synaptic and memory deficits in a transgenic mouse model of α-synucleinopathy. Acta Neuropathol 2019; 138:551-574. [PMID: 31168644 PMCID: PMC6778173 DOI: 10.1007/s00401-019-02032-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/27/2019] [Accepted: 05/27/2019] [Indexed: 01/01/2023]
Abstract
Parkinson's disease dementia (PDD) and dementia with Lewy bodies (DLB) are clinically and neuropathologically highly related α-synucleinopathies that collectively constitute the second leading cause of neurodegenerative dementias. Genetic and neuropathological studies directly implicate α-synuclein (αS) abnormalities in PDD and DLB pathogenesis. However, it is currently unknown how αS abnormalities contribute to memory loss, particularly since forebrain neuronal loss in PDD and DLB is less severe than in Alzheimer's disease. Previously, we found that familial Parkinson's disease-linked human mutant A53T αS causes aberrant localization of the microtubule-associated protein tau to postsynaptic spines in neurons, leading to postsynaptic deficits. Thus, we directly tested if the synaptic and memory deficits in a mouse model of α-synucleinopathy (TgA53T) are mediated by tau. TgA53T mice exhibit progressive memory deficits associated with postsynaptic deficits in the absence of obvious neuropathological and neurodegenerative changes in the hippocampus. Significantly, removal of endogenous mouse tau expression in TgA53T mice (TgA53T/mTau-/-), achieved by mating TgA53T mice to mouse tau-knockout mice, completely ameliorates cognitive dysfunction and concurrent synaptic deficits without affecting αS expression or accumulation of selected toxic αS oligomers. Among the known tau-dependent effects, memory deficits in TgA53T mice were associated with hippocampal circuit remodeling linked to chronic network hyperexcitability. This remodeling was absent in TgA53T/mTau-/- mice, indicating that postsynaptic deficits, aberrant network hyperactivity, and memory deficits are mechanistically linked. Our results directly implicate tau as a mediator of specific human mutant A53T αS-mediated abnormalities related to deficits in hippocampal neurotransmission and suggest a mechanism for memory impairment that occurs as a consequence of synaptic dysfunction rather than synaptic or neuronal loss. We hypothesize that these initial synaptic deficits contribute to network hyperexcitability which, in turn, exacerbate cognitive dysfunction. Our results indicate that these synaptic changes present potential therapeutic targets for amelioration of memory deficits in α-synucleinopathies.
Collapse
Affiliation(s)
- Balvindar Singh
- Medical Scientist Training Program, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
- Graduate Program in Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Ana Covelo
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Héctor Martell-Martínez
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Carmen Nanclares
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Mathew A Sherman
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Emmanuel Okematti
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Joyce Meints
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Peter J Teravskis
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Christopher Gallardo
- Graduate Program in Pharmacology, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Alena V Savonenko
- Department of Pathology, Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD, 21205, USA
| | - Michael A Benneyworth
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
- Institute for Translational Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
- Mouse Behavior Core, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Sylvain E Lesné
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
- Institute for Translational Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
- N. Budd Grossman Center for Memory Research and Care, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Dezhi Liao
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
- Institute for Translational Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Alfonso Araque
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
- Institute for Translational Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Michael K Lee
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA.
- Institute for Translational Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA.
- Geriatric Research Education and Clinical Center, Minneapolis Veterans Affairs Health Care System, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
227
|
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: 7.8] [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
|
228
|
Lam AD, Cole AJ, Cash SS. New Approaches to Studying Silent Mesial Temporal Lobe Seizures in Alzheimer's Disease. Front Neurol 2019; 10:959. [PMID: 31551916 PMCID: PMC6737997 DOI: 10.3389/fneur.2019.00959] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/20/2019] [Indexed: 12/13/2022] Open
Abstract
Silent seizures were discovered in mouse models of Alzheimer's disease over 10 years ago, yet it remains unclear whether these seizures are a salient feature of Alzheimer's disease in humans. Seizures that arise early in the course of Alzheimer's disease most likely originate from the mesial temporal lobe, one of the first structures affected by Alzheimer's disease pathology and one of the most epileptogenic regions of the brain. Several factors greatly limit our ability to identify mesial temporal lobe seizures in patients with Alzheimer's disease, however. First, mesial temporal lobe seizures can be difficult to recognize clinically, as their accompanying symptoms are often subtle or even non-existent. Second, electrical activity arising from the mesial temporal lobe is largely invisible on the scalp electroencephalogram (EEG), the mainstay of diagnosis for epilepsy in this population. In this review, we will describe two new approaches being used to study silent mesial temporal lobe seizures in Alzheimer's disease. We will first describe the methodology and application of foramen ovale electrodes, which captured the first recordings of silent mesial temporal lobe seizures in humans with Alzheimer's disease. We will then describe machine learning approaches being developed to non-invasively identify silent mesial temporal lobe seizures on scalp EEG. Both of these tools have the potential to elucidate the role of silent seizures in humans with Alzheimer's disease, which could have important implications for early diagnosis, prognostication, and development of targeted therapies for this population.
Collapse
Affiliation(s)
- Alice D. Lam
- Massachusetts General Hospital, Department of Neurology, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Andrew J. Cole
- Massachusetts General Hospital, Department of Neurology, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Sydney S. Cash
- Massachusetts General Hospital, Department of Neurology, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| |
Collapse
|
229
|
Qin Q, Li Y. Herpesviral infections and antimicrobial protection for Alzheimer's disease: Implications for prevention and treatment. J Med Virol 2019; 91:1368-1377. [PMID: 30997676 DOI: 10.1002/jmv.25481] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 04/04/2019] [Indexed: 02/05/2023]
Abstract
Accumulating evidence suggests that infections by herpesviruses might be closely linked to Alzheimer's disease (AD). Pathological hallmarks of AD brains include senile plaques induced by amyloid β peptide (Aβ) in the extracellular space and intracellular neurofibrillary tangles (NFTs) consisting of phosphorylated tau protein. The prevailing hypothesis for the mechanism of AD is amyloid cascade reaction. Recent studies revealed that infections by herpesviruses induce the similar pathological hallmarks of AD, including Aβ production, phosphorylation of tau (P-tau), oxidative stress, neuroinflammation, etc. Aβ peptide is regarded as one of the antimicrobial peptides, which inhibits HSV-1 replication. In the elderly, reactivation of herpesviruses might act as an initiator for amyloid cascade reaction in vulnerable individuals, triggering the neurofibrillary formation of phosphorylated tau and inducing oxidative stress and neuroinflammation, which can further contribute to the accumulation of Aβ and P-tau by impairing mitochondria and autophagosome. Epidemiological studies have shown AD susceptibility genes, such as APOE-ε4 allele, are highly linked to infections by herpesviruses. Interestingly, anti-herpesviral therapy significantly reduced the risk of AD in a large population study. Given that herpesviruses are arguably the most prevalent opportunistic pathogens and often reactivate in the elderly, it is reasonable to argue reactivation of herpesviruses might be major culprits for initiating AD in individuals carrying AD susceptibility genes. In this review, we summarize epidemiological and molecular evidence that support for a hypothesis of herpesviral infections and antimicrobial protection in the development of AD, and discuss the implications for future prevention and treatment of the disease.
Collapse
Affiliation(s)
- Qingsong Qin
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou, Guangdong, China
| | - Yun Li
- Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong, China
- Mental Health Center, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
230
|
Inaba T, Miyamoto N, Hira K, Ueno Y, Yamashiro K, Watanabe M, Shimada Y, Hattori N, Urabe T. Protective Role of Levetiracetam Against Cognitive Impairment And Brain White Matter Damage in Mouse prolonged Cerebral Hypoperfusion. Neuroscience 2019; 414:255-264. [PMID: 31302262 DOI: 10.1016/j.neuroscience.2019.07.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 10/26/2022]
Abstract
White matter lesions due to cerebral hypoperfusion may be an important pathophysiology in vascular dementia and stroke, although the inherent mechanisms remain to be fully elucidated. The present study, using a mouse model of chronic cerebral hypoperfusion, examined the white matter protective effects of levetiracetam, an anticonvulsant, via the signaling cascade from the activation of cAMP-responsive element binding protein (CREB) phosphorylation. Mice underwent bilateral common carotid artery stenosis (BCAS), and were separated into the levetiracetam group (injected once only after BCAS [LEV1] or injected on three consecutive days [LEV3]), the vehicle group, or the anti-epileptic drugs with different action mechanisms phenytoin group (PHT3; injected on three consecutive days with the same condition as in LEV3). Cerebral blood flow analysis, Y-maze spontaneous alternation test, novel object recognition test, immunohistochemical and Western blot analyses, and protein kinase A assay were performed after BCAS. In the LEV3 group, SV2A expression was markedly increased, which preserved learning and memory after BCAS. Moreover, as the protein kinase A level was significantly increased, pCREB expression was also increased. The activation of microglia and astrocytes was markedly suppressed, although the number of oligodendrocyte precursor cells (OPCs) and GST-pi-positive-oligodendrocytes was markedly higher in the cerebral white matter. Moreover, oxidative stress was significantly reduced. We found that 3-day treatment with levetiracetam maintained SV2A protein expression via interaction with astrocytes, which influenced the OPC lineage through activation of CREB to protect white matter from ischemia.
Collapse
Affiliation(s)
- Toshiki Inaba
- Department of Neurology, Juntendo University Urayasu Hospital, Chiba, Japan; Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Nobukazu Miyamoto
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan.
| | - Kenichiro Hira
- Department of Neurology, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Yuji Ueno
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Kazuo Yamashiro
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Masao Watanabe
- Department of Neurology, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Yoshiaki Shimada
- Department of Neurology, Juntendo University Urayasu Hospital, Chiba, Japan; Department of Neurological Science, Yokohama Tsurugamine Hospital, Yokohama, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Takao Urabe
- Department of Neurology, Juntendo University Urayasu Hospital, Chiba, Japan
| |
Collapse
|
231
|
Frere S, Slutsky I. Alzheimer's Disease: From Firing Instability to Homeostasis Network Collapse. Neuron 2019; 97:32-58. [PMID: 29301104 DOI: 10.1016/j.neuron.2017.11.028] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/14/2017] [Accepted: 11/17/2017] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) starts from pure cognitive impairments and gradually progresses into degeneration of specific brain circuits. Although numerous factors initiating AD have been extensively studied, the common principles underlying the transition from cognitive deficits to neuronal loss remain unknown. Here we describe an evolutionarily conserved, integrated homeostatic network (IHN) that enables functional stability of central neural circuits and safeguards from neurodegeneration. We identify the critical modules comprising the IHN and propose a central role of neural firing in controlling the complex homeostatic network at different spatial scales. We hypothesize that firing instability and impaired synaptic plasticity at early AD stages trigger a vicious cycle, leading to dysregulation of the whole IHN. According to this hypothesis, the IHN collapse represents the major driving force of the transition from early memory impairments to neurodegeneration. Understanding the core elements of homeostatic control machinery, the reciprocal connections between distinct IHN modules, and the role of firing homeostasis in this hierarchy has important implications for physiology and should offer novel conceptual approaches for AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Samuel Frere
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel.
| |
Collapse
|
232
|
Coelho DS, Moreno E. Emerging links between cell competition and Alzheimer's disease. J Cell Sci 2019; 132:132/13/jcs231258. [PMID: 31263078 DOI: 10.1242/jcs.231258] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Alzheimer's disease (AD) causes a progressive loss of memory and other cognitive functions, which inexorably debilitates patients. There is still no cure for AD and effective treatments to delay or revert AD are urgently needed. On a molecular level, the excessive accumulation of amyloid-β (Aβ) peptides triggers a complex cascade of pathological events underlying neuronal death, whose details are not yet completely understood. Our laboratory recently discovered that cell competition may play a protective role against AD by eliminating less fit neurons from the brain of Aβ-transgenic flies. Loss of Aβ-damaged neurons through fitness comparison with healthy counterparts is beneficial for the organism, delaying cognitive decline and motor disability. In this Review, we introduce the molecular mechanisms of cell competition, including seminal works on the field and latest advances regarding genetic triggers and effectors of cell elimination. We then describe the biological relevance of competition in the nervous system and discuss how competitive interactions between neurons may arise and be exacerbated in the context of AD. Selection of neurons through fitness comparison is a promising, but still emerging, research field that may open new avenues for the treatment of neurological disorders.
Collapse
Affiliation(s)
- Dina S Coelho
- Cell Fitness Laboratory, Champalimaud Centre for the Unknown, Av. Brasília., 1400-038 Lisbon, Portugal
| | - Eduardo Moreno
- Cell Fitness Laboratory, Champalimaud Centre for the Unknown, Av. Brasília., 1400-038 Lisbon, Portugal
| |
Collapse
|
233
|
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: 9.5] [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
|
234
|
Powell G, Ziso B, Larner AJ. The overlap between epilepsy and Alzheimer's disease and the consequences for treatment. Expert Rev Neurother 2019; 19:653-661. [PMID: 31238746 DOI: 10.1080/14737175.2019.1629289] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Introduction: Alzheimer's disease may be associated with both clinical and subclinical epileptic seizure activity. Once regarded as an epiphenomenon, epileptiform activity may, in fact, be an integral part of the Alzheimer's phenotype, and may be not only a symptomatic therapeutic target but also a possible mechanism to retard or prevent disease progression. Areas covered: The authors review clinical research articles with a focus on the semiology, epidemiology, and treatment of seizures in Alzheimer's disease, and also look at some experimental animal model studies which have informed clinical thinking on seizure aetiopathogenesis. The evidence base for treatment decisions is sparse. A brief overview of the clinical assessment of Alzheimer's disease patients considering relevant differential diagnoses and diagnostic pitfalls is presented. Expert opinion: Studies of epileptic seizures in Alzheimer's disease have become more frequent over the last 5-10 years. Understanding of seizure semiology, epidemiology, and possible pathogenesis has increased. However, the optimal management of seizures in this context remains unknown, largely due to the paucity of studies sufficient to examine this question. Clearly, such studies will be required, not only to inform clinicians about symptomatic control of seizures in Alzheimer's disease but also to investigate whether this might impact on disease progression.
Collapse
Affiliation(s)
- Graham Powell
- a Mersey Regional Epilepsy Clinic , Walton Centre for Neurology and Neurosurgery , Liverpool , UK
| | - Besa Ziso
- a Mersey Regional Epilepsy Clinic , Walton Centre for Neurology and Neurosurgery , Liverpool , UK
| | - A J Larner
- b Cognitive Function Clinic , Walton Centre for Neurology and Neurosurgery , Liverpool , UK
| |
Collapse
|
235
|
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: 9.5] [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
|
236
|
Henderson BW, Greathouse KM, Ramdas R, Walker CK, Rao TC, Bach SV, Curtis KA, Day JJ, Mattheyses AL, Herskowitz JH. Pharmacologic inhibition of LIMK1 provides dendritic spine resilience against β-amyloid. Sci Signal 2019; 12:eaaw9318. [PMID: 31239325 PMCID: PMC7088434 DOI: 10.1126/scisignal.aaw9318] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) therapies predominantly focus on β-amyloid (Aβ), but Aβ effects may be maximal before clinical symptoms appear. Downstream of Aβ, dendritic spine loss correlates most strongly with cognitive decline in AD. Rho-associated kinases (ROCK1 and ROCK2) regulate the actin cytoskeleton, and ROCK1 and ROCK2 protein abundances are increased in early AD. Here, we found that the increased abundance of ROCK1 in cultured primary rat hippocampal neurons reduced dendritic spine length through a myosin-based pathway, whereas the increased abundance of ROCK2 induced spine loss through the serine and threonine kinase LIMK1. Aβ42 oligomers can activate ROCKs. Here, using static imaging studies combined with multielectrode array analyses, we found that the ROCK2-LIMK1 pathway mediated Aβ42-induced spine degeneration and neuronal hyperexcitability. Live-cell microscopy revealed that pharmacologic inhibition of LIMK1 rendered dendritic spines resilient to Aβ42 oligomers. Treatment of hAPP mice with a LIMK1 inhibitor rescued Aβ-induced hippocampal spine loss and morphologic aberrations. Our data suggest that therapeutically targeting LIMK1 may provide dendritic spine resilience to Aβ and therefore may benefit cognitively normal patients that are at high risk for developing dementia.
Collapse
Affiliation(s)
- Benjamin W Henderson
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
- Department of Neurology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Kelsey M Greathouse
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
- Department of Neurology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Raksha Ramdas
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
- Department of Neurology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Courtney K Walker
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
- Department of Neurology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Tejeshwar C Rao
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Svitlana V Bach
- Department of Neurobiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Kendall A Curtis
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
- Department of Neurology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Jeremy J Day
- Department of Neurobiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Alexa L Mattheyses
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Jeremy H Herskowitz
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA.
- Department of Neurology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| |
Collapse
|
237
|
Edwards III GA, Gamez N, Escobedo Jr. G, Calderon O, Moreno-Gonzalez I. Modifiable Risk Factors for Alzheimer's Disease. Front Aging Neurosci 2019; 11:146. [PMID: 31293412 PMCID: PMC6601685 DOI: 10.3389/fnagi.2019.00146] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 05/31/2019] [Indexed: 01/03/2023] Open
Abstract
Since first described in the early 1900s, Alzheimer's disease (AD) has risen exponentially in prevalence and concern. Research still drives to understand the etiology and pathogenesis of this disease and what risk factors can attribute to AD. With a majority of AD cases being of sporadic origin, the increasing exponential growth of an aged population and a lack of treatment, it is imperative to discover an easy accessible preventative method for AD. Some risk factors can increase the propensity of AD such as aging, sex, and genetics. Moreover, there are also modifiable risk factors-in terms of treatable medical conditions and lifestyle choices-that play a role in developing AD. These risk factors have their own biological mechanisms that may contribute to AD etiology and pathological consequences. In this review article, we will discuss modifiable risk factors and discuss the current literature of how each of these factors interplay into AD development and progression and if strategically analyzed and treated, could aid in protection against this neurodegenerative disease.
Collapse
Affiliation(s)
- George A. Edwards III
- The Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, The University of Texas Houston Health Science Center at Houston, Houston, TX, United States
| | - Nazaret Gamez
- The Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, The University of Texas Houston Health Science Center at Houston, Houston, TX, United States
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Department of Cell Biology, Facultad Ciencias, Universidad de Malaga, Malaga, Spain
| | - Gabriel Escobedo Jr.
- The Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, The University of Texas Houston Health Science Center at Houston, Houston, TX, United States
| | - Olivia Calderon
- The Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, The University of Texas Houston Health Science Center at Houston, Houston, TX, United States
| | - Ines Moreno-Gonzalez
- The Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, The University of Texas Houston Health Science Center at Houston, Houston, TX, United States
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Department of Cell Biology, Facultad Ciencias, Universidad de Malaga, Malaga, Spain
| |
Collapse
|
238
|
Wang MJ, Jiang L, Chen HS, Cheng L. Levetiracetam Protects Against Cognitive Impairment of Subthreshold Convulsant Discharge Model Rats by Activating Protein Kinase C (PKC)-Growth-Associated Protein 43 (GAP-43)-Calmodulin-Dependent Protein Kinase (CaMK) Signal Transduction Pathway. Med Sci Monit 2019; 25:4627-4638. [PMID: 31266934 PMCID: PMC6601366 DOI: 10.12659/msm.913542] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Subclinical epileptiform discharges (SEDs) are defined as epileptiform electroencephalographic (EEG) discharges without clinical signs of seizure in patients. The subthreshold convulsant discharge (SCD) is a frequently used model for SEDs. This study aimed to investigate the effect of levetiracetam (LEV), an anti-convulsant drug, on cognitive impairment of SCD model rats and to assess the associated mechanisms. Material/Methods A SCD rat model was established. Rats were divided into an SCD group, an SCD+ sodium valproate (VPA) group, and an SCD+ levetiracetam (LEV) group. The Morris water maze was used to evaluate the capacity of positioning navigation and space exploration. The field excitatory post-synaptic potentials (fEPSPs) were evaluated using a bipolar stimulation electrode. NCAM, GAP43, PS95, and CaMK II levels were detected using Western blot and RT-PCR, respectively. PKC activity was examined by a non-radioactive method. Results LEV shortens the latency of platform seeking in SCD rats in positioning navigation. fEPSP slopes were significantly lower in the SCD group, and LEV treatment significantly enhanced the fEPSP slopes compared to the SCD group (P<0.05). The NCAM and GAP-43 levels were increased and PSD-95 levels were increased in SCD rats (P<0.05), which were improved by LEV treatment. The PKC activity and CaMK II levels were decreased in SCD rats and LEV treatment significantly enhanced PKC activity and increased CaMK II levels. Conclusions Cognitive impairment in of SCD model rats may be caused by decreased PKC activity, low expression of CaMK II, and inhibition of LTP formation. LEV can improve cognitive function by activating the PKC-GAP-43-CaMK signal transduction pathway.
Collapse
Affiliation(s)
- Min-Jian Wang
- Department of Psychology, Children's Hospital of Chongqing Medical University, Chongqing, China (mainland).,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China (mainland).,Key Laboratory of Pediatrics in Chongqing, Chongqing, China (mainland).,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China (mainland)
| | - Li Jiang
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China (mainland).,Key Laboratory of Pediatrics in Chongqing, Chongqing, China (mainland).,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China (mainland).,Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Heng-Sheng Chen
- Key Laboratory of Pediatrics in Chongqing, Chongqing, China (mainland).,Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Li Cheng
- Key Laboratory of Pediatrics in Chongqing, Chongqing, China (mainland).,Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, China (mainland)
| |
Collapse
|
239
|
Herbal Formula Fo Shou San Attenuates Alzheimer's Disease-Related Pathologies via the Gut-Liver-Brain Axis in APP/PS1 Mouse Model of Alzheimer's Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:8302950. [PMID: 31316576 PMCID: PMC6601474 DOI: 10.1155/2019/8302950] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 05/20/2019] [Indexed: 02/07/2023]
Abstract
Fo Shou San (FSS) is an ancient paired-herb decoction, used in China to treat blood deficiency, blood stasis, stroke, and ischemic cerebral vascular disease for about one thousand years. The mechanisms associated with these properties, however, are not completely understood. Gut bacteria, gut bacterial lipopolysaccharides (LPS), alkaline phosphatase (AP), and lipid peroxidation are common biochemical signaling that takes place on gut-liver-brain axis. Growing evidences have revealed that gut bacterial lipopolysaccharides (LPS) enter the systemic circulation via the portal vein, and finally entering the brain tissue is an important cause of inflammatory degeneration of Alzheimer's disease (AD). Alkaline phosphatase (AP) dephosphorylates LPS forming a nontoxic LPS and reduces systemic inflammation via gut-liver-brain axis. In this study, to identify the differentially gut-liver-brain axis among APP/PS1 mice, FSS-treated APP/PS1 mice, and control mice, behavioral tests were performed to assess the cognitive ability and hematoxylin-eosin staining was used to assess neuronal damage in the hippocampus; immunohistochemistry, western blotting, a quantitative chromogenic end-point Tachypleus amebocyte lysate (TAL) assay kit, Malondialdehyde (MDA) assay kit, AP Assay Kit, and real-time quantitative PCR (qPCR) were used to assess the level of LPS, MDA, AP, and gut bacteria. We found that FSS regulates gut-liver-brain axis to regulate AP and gut bacteria and attenuate the LPS-related systemic inflammation, oxidative stress (MDA), and thereby AD-related pathology in APP/PS1 mice. This is the first study to provide a reference for FSS-treated AD mice to aid in understanding the underlying mechanisms of FSS. FSS may also improve gastrointestinal tract barrier and blood-brain barrier and thus ameliorates the symptoms of AD; this is subject to our further study.
Collapse
|
240
|
Zott B, Busche MA, Sperling RA, Konnerth A. What Happens with the Circuit in Alzheimer's Disease in Mice and Humans? Annu Rev Neurosci 2019; 41:277-297. [PMID: 29986165 DOI: 10.1146/annurev-neuro-080317-061725] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A major mystery of many types of neurological and psychiatric disorders, such as Alzheimer's disease (AD), remains the underlying, disease-specific neuronal damage. Because of the strong interconnectivity of neurons in the brain, neuronal dysfunction necessarily disrupts neuronal circuits. In this article, we review evidence for the disruption of large-scale networks from imaging studies of humans and relate it to studies of cellular dysfunction in mouse models of AD. The emerging picture is that some forms of early network dysfunctions can be explained by excessively increased levels of neuronal activity. The notion of such neuronal hyperactivity receives strong support from in vivo and in vitro cellular imaging and electrophysiological recordings in the mouse, which provide mechanistic insights underlying the change in neuronal excitability. Overall, some key aspects of AD-related neuronal dysfunctions in humans and mice are strikingly similar and support the continuation of such a translational strategy.
Collapse
Affiliation(s)
- Benedikt Zott
- Institute of Neuroscience, Technical University of Munich, 80802 Munich, Germany; .,Center for Integrated Protein Sciences, Technical University of Munich, 80802 Munich, Germany.,Munich Cluster for Systems Neurology, Technical University of Munich, 80802 Munich, Germany
| | - Marc Aurel Busche
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA.,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA.,Department of Neurology and Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Arthur Konnerth
- Institute of Neuroscience, Technical University of Munich, 80802 Munich, Germany; .,Center for Integrated Protein Sciences, Technical University of Munich, 80802 Munich, Germany.,Munich Cluster for Systems Neurology, Technical University of Munich, 80802 Munich, Germany
| |
Collapse
|
241
|
Najm R, Jones EA, Huang Y. Apolipoprotein E4, inhibitory network dysfunction, and Alzheimer's disease. Mol Neurodegener 2019; 14:24. [PMID: 31186040 PMCID: PMC6558779 DOI: 10.1186/s13024-019-0324-6] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 05/23/2019] [Indexed: 02/08/2023] Open
Abstract
Apolipoprotein (apo) E4 is the major genetic risk factor for Alzheimer's disease (AD), increasing risk and decreasing age of disease onset. Many studies have demonstrated the detrimental effects of apoE4 in varying cellular contexts. However, the underlying mechanisms explaining how apoE4 leads to cognitive decline are not fully understood. Recently, the combination of human induced pluripotent stem cell (hiPSC) modeling of neurological diseases in vitro and electrophysiological studies in vivo have begun to unravel the intersection between apoE4, neuronal subtype dysfunction or loss, subsequent network deficits, and eventual cognitive decline. In this review, we provide an overview of the literature describing apoE4's detrimental effects in the central nervous system (CNS), specifically focusing on its contribution to neuronal subtype dysfunction or loss. We focus on γ-aminobutyric acid (GABA)-expressing interneurons in the hippocampus, which are selectively vulnerable to apoE4-mediated neurotoxicity. Additionally, we discuss the importance of the GABAergic inhibitory network to proper cognitive function and how dysfunction of this network manifests in AD. Finally, we examine how apoE4-mediated GABAergic interneuron loss can lead to inhibitory network deficits and how this deficit results in cognitive decline. We propose the following working model: Aging and/or stress induces neuronal expression of apoE. GABAergic interneurons are selectively vulnerable to intracellularly produced apoE4, through a tau dependent mechanism, which leads to their dysfunction and eventual death. In turn, GABAergic interneuron loss causes hyperexcitability and dysregulation of neural networks in the hippocampus and cortex. This dysfunction results in learning, memory, and other cognitive deficits that are the central features of AD.
Collapse
Affiliation(s)
- Ramsey Najm
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, CA, 94143, USA
| | - Emily A Jones
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, 94143, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA.
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, CA, 94143, USA.
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, 94143, USA.
- Department of Neurology, University of California, San Francisco, CA, 94143, USA.
- Department of Pathology, University of California, San Francisco, CA, 94143, USA.
| |
Collapse
|
242
|
Linsley JW, Reisine T, Finkbeiner S. Cell death assays for neurodegenerative disease drug discovery. Expert Opin Drug Discov 2019; 14:901-913. [PMID: 31179783 DOI: 10.1080/17460441.2019.1623784] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: Neurodegenerative diseases affect millions of people worldwide. Neurodegeneration is gradual over time, characterized by neuronal death that causes deterioration of cognitive or motor functions, ultimately leading to the patient's death. Currently, there are no treatments that effectively slow the progression of any neurodegenerative disease, but improved microscopy assays and models for neurodegeneration could lead the way to the discovery of disease-modifying therapeutics. Areas covered: Herein, the authors describe cell-based assays used to discover drugs with the potential to slow neurodegeneration, and their associated disease models. They focus on microscopy technologies that can be adapted to a high-throughput screening format that both detect cell death and monitor early signs of neurodegeneration and functional changes to identify drugs that the block early stages of neurodegeneration. Expert opinion: Many different phenotypes have been used in screens for the development of therapeutics towards neurodegenerative disease. The context of each phenotype in relation to neurodegeneration must be established to identify therapeutics likely to successfully target and treat disease. The use of improved models of neurodegeneration, statistical analyses, computational models, and improved markers of neuronal death will help in this pursuit and lead to better screening methods to identify therapeutic compounds against neurodegenerative disease.
Collapse
Affiliation(s)
- Jeremy W Linsley
- a Gladstone Center for Systems and Therapeutics , San Francisco , CA , USA
| | - Terry Reisine
- b Independent scientific consultant , Santa Cruz , CA , USA
| | - Steven Finkbeiner
- a Gladstone Center for Systems and Therapeutics , San Francisco , CA , USA.,c Neuroscience Graduate Program, University of California , San Francisco , CA , USA.,d Biomedical Sciences and Neuroscience Graduate Program, University of California , San Francisco , CA , USA.,e Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes , San Francisco , CA , USA.,f Department of Neurology, University of California , San Francisco , CA , USA.,g Department of Physiology, University of California , San Francisco , CA , USA
| |
Collapse
|
243
|
Shu H, Shi Y, Chen G, Wang Z, Liu D, Yue C, Ward BD, Li W, Xu Z, Chen G, Guo QH, Xu J, Li SJ, Zhang Z. Distinct neural correlates of episodic memory among apolipoprotein E alleles in cognitively normal elderly. Brain Imaging Behav 2019; 13:255-269. [PMID: 29396739 DOI: 10.1007/s11682-017-9818-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The apolipoprotein E (APOE) ε4 and ε2 alleles are acknowledged genetic factors modulating Alzheimer's disease (AD) risk and episodic memory (EM) deterioration in an opposite manner. Mounting neuroimaging studies describe EM-related brain activity differences among APOE alleles but remain limited in elucidating the underlying mechanism. Here, we hypothesized that the APOE ε2, ε3, and ε4 alleles have distinct EM neural substrates, as a manifestation of degeneracy, underlying their modulations on EM-related brain activity and AD susceptibility. To test the hypothesis, we identified neural correlates of EM function by correlating intrinsic hippocampal functional connectivity networks with neuropsychological EM performances in a voxelwise manner, with 129 cognitively normal elderly subjects (36 ε2 carriers, 44 ε3 homozygotes, and 49 ε4 carriers). We demonstrated significantly different EM neural correlates among the three APOE allele groups. Specifically, in the ε3 homozygotes, positive EM neural correlates were characterized in the Papez circuit regions; in the ε4 carriers, positive EM neural correlates involved the lateral temporal cortex, premotor cortex/sensorimotor cortex/superior parietal lobule, and cuneus; and in the ε2 carriers, negative EM neural correlates appeared in the bilateral frontopolar, posteromedial, and sensorimotor cortex. Further, in the ε4 carriers, the interaction between age and EM function occurred in the temporoparietal junction and prefrontal cortex. Our findings suggest that the underlying mechanism of APOE polymorphism modulations on EM function and AD susceptibility is genetically related to the neural degeneracy of EM function across APOE alleles.
Collapse
Affiliation(s)
- Hao Shu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu, 210009, China
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Yongmei Shi
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu, 210009, China
| | - Gang Chen
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Zan Wang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu, 210009, China
| | - Duan Liu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu, 210009, China
| | - Chunxian Yue
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu, 210009, China
| | - B Douglas Ward
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Wenjun Li
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Zhan Xu
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Guangyu Chen
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Qi-Hao Guo
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jun Xu
- Department of Neurology, Jiangsu Province Geriatric Institute, Nanjing, Jiangsu, 210024, China
| | - Shi-Jiang Li
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| | - Zhijun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu, 210009, China.
| |
Collapse
|
244
|
Synthesis and in vivo evaluation of [ 18F]UCB-J for PET imaging of synaptic vesicle glycoprotein 2A (SV2A). Eur J Nucl Med Mol Imaging 2019; 46:1952-1965. [PMID: 31175396 DOI: 10.1007/s00259-019-04357-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/02/2019] [Indexed: 02/05/2023]
Abstract
PURPOSE Synaptic abnormalities have been implicated in a variety of neuropsychiatric disorders, including epilepsy, Alzheimer's disease, and schizophrenia. Hence, PET imaging of the synaptic vesicle glycoprotein 2A (SV2A) may be a valuable in vivo biomarker for neurologic and psychiatric diseases. We previously developed [11C]UCB-J, a PET radiotracer with high affinity and selectivity toward SV2A; however, the short radioactive half-life (20 min for 11C) places some limitations on its broader application. Herein, we report the first synthesis of the longer-lived 18F-labeled counterpart (half-life: 110 min), [18F]UCB-J, and its evaluation in nonhuman primates. METHODS [18F]UCB-J was synthesized from the iodonium precursors. PET imaging experiments with [18F]UCB-J were conducted in rhesus monkeys to assess the pharmacokinetic and in vivo binding properties. Arterial samples were taken for analysis of radioactive metabolites and generation of input functions. Regional time-activity curves were analyzed using the one-tissue compartment model to derive regional distribution volumes and binding potentials for comparison with [11C]UCB-J. RESULTS [18F]UCB-J was prepared in high radiochemical and enantiomeric purity, but low radiochemical yield. Evaluation in nonhuman primates indicated that the radiotracer displayed pharmacokinetic and imaging characteristics similar to those of [11C]UCB-J, with moderate metabolism rate, high brain uptake, fast and reversible binding kinetics, and high specific binding signals. CONCLUSION We have accomplished the first synthesis of the novel SV2A radiotracer [18F]UCB-J. [18F]UCB-J is demonstrated to be an excellent imaging agent and may prove to be useful for imaging and quantification of SV2A expression, and synaptic density, in humans.
Collapse
|
245
|
Wang C, Cai Z, Wang W, Wei M, Kou D, Li T, Yang Z, Guo H, Le W, Li S. Piperine attenuates cognitive impairment in an experimental mouse model of sporadic Alzheimer's disease. J Nutr Biochem 2019; 70:147-155. [PMID: 31207354 DOI: 10.1016/j.jnutbio.2019.05.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/25/2019] [Accepted: 05/14/2019] [Indexed: 12/20/2022]
Abstract
Piperine, the major alkaloid constituent of black pepper, has been reported to possess a wide range of pharmacological effects on the central nervous system, including antidepressant, anticonvulsant and anti-ischemic activities. In the present study, we aimed to investigate the therapeutic potential and neuroprotective mechanisms of piperine in an experimental mouse model of sporadic Alzheimer's disease (sAD) induced by intracerebroventricular (ICV) infusion of streptozotocin (STZ). STZ was infused bilaterally at a dose of 1.5 mg/kg/day on day 1 and day 3. From day 8, piperine (2.5-10 mg/kg body weight) was administered intraperitoneally once daily for 15 consecutive days. The locomotor activity and cognitive performance of mice were evaluated using open field test and Morris water maze test, respectively. On day 23, all animals were sacrificed, and the hippocampus was used for biochemical, neurochemical and neuroinflammatory determinations. Our data revealed that the ICV-STZ-infused sAD mouse showed an increased oxidative-nitrosative stress, an altered neurotransmission and an elevated neuroinflammation in hippocampus, as well as significant cognitive deficits. All these alterations can be ameliorated by piperine in a dose-dependent manner. In summary, our findings predict a therapeutic potential of piperine against cognitive deficits in sAD mouse. This effect might be due to its abilities to ameliorate oxidative-nitrosative stress, restore neurotransmission and reduce neuroinflammation.
Collapse
Affiliation(s)
- Che Wang
- Department of Medicinal Chemistry, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Zhengxu Cai
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian 116011, China
| | - Wei Wang
- Department of Rehabilitation, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Min Wei
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116011, China; Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116011, China
| | - Daqing Kou
- Department of Clinical Laboratory, the First Affiliated Hospital, Dalian Medical University, Dalian 116011, China
| | - Tianbai Li
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116011, China; Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116011, China
| | - Zhaofei Yang
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116011, China; Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116011, China
| | - Huishu Guo
- Central Laboratory, the First Affiliated Hospital, Dalian Medical University, Dalian 116021, China.
| | - Weidong Le
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116011, China; Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116011, China.
| | - Song Li
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116011, China; Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
246
|
Ciruelas K, Marcotulli D, Bajjalieh SM. Synaptic vesicle protein 2: A multi-faceted regulator of secretion. Semin Cell Dev Biol 2019; 95:130-141. [PMID: 30826548 DOI: 10.1016/j.semcdb.2019.02.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/11/2019] [Accepted: 02/21/2019] [Indexed: 01/01/2023]
Abstract
Synaptic Vesicle Protein 2 (SV2) comprises a recently evolved family of proteins unique to secretory vesicles that undergo calcium-regulated exocytosis. In this review we consider SV2s' structural features, evolution, and function and discuss its therapeutic potential as the receptors for an expanding class of drugs used to treat epilepsy and cognitive decline.
Collapse
Affiliation(s)
- Kristine Ciruelas
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Daniele Marcotulli
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Sandra M Bajjalieh
- Department of Pharmacology, University of Washington, Seattle, WA, United States.
| |
Collapse
|
247
|
Popiolek M, Mandelblat-Cerf Y, Young D, Garst-Orozco J, Lotarski SM, Stark E, Kramer M, Butler CR, Kozak R. In Vivo Modulation of Hippocampal Excitability by M4 Muscarinic Acetylcholine Receptor Activator: Implications for Treatment of Alzheimer's Disease and Schizophrenic Patients. ACS Chem Neurosci 2019; 10:1091-1098. [PMID: 30335349 DOI: 10.1021/acschemneuro.8b00496] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Abnormal hippocampal activity has been linked to impaired cognitive performance in Alzheimer's disease and schizophrenia, leading to a hypothesis that normalization of this activity may be therapeutically beneficial. Our work suggests that one approach for hippocampal normalization may be through activation of the M4 muscarinic acetylcholine receptor. We used a brain penetrant M4 muscarinic acetylcholine receptor selective activator, PT-3763, to show dose-dependent attenuation of field potentials in Schaffer collateral (CA3-CA1) and recurrent associational connections (CA3-CA3) ex vivo in hippocampal slices. In vivo, systemic administration of PT-3763 led to attenuation of glutamate release in CA3 as measured by amperometry and to a dose-dependent decrease in population CA1 pyramidal activity as measured by fiber photometry. This decrease in population activity was also evident with a localized administration of the compound to the recorded site. Finally, PT-3763 reversed scopolamine-induced deficit in Morris water maze. Our results suggest that M4 muscarinic acetylcholine receptor activation may be a suitable therapeutic treatment in diseases associated with hyperactive hippocampal activity.
Collapse
|
248
|
Dewey CW, Davies ES, Xie H, Wakshlag JJ. Canine Cognitive Dysfunction: Pathophysiology, Diagnosis, and Treatment. Vet Clin North Am Small Anim Pract 2019; 49:477-499. [PMID: 30846383 DOI: 10.1016/j.cvsm.2019.01.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Canine cognitive dysfunction (CCD) is the canine analog of human Alzheimer disease (AD). The pathophysiology of CCD/AD is multifaceted. CCD is common in aged (>8 years) dogs, affecting between 14% and 35% of the pet dog population. Apparent confusion, anxiety, disturbance of the sleep/wake cycle, and decreased interaction with owners are all common clinical signs of CCD. Although there is no cure for CCD, several proven effective therapeutic approaches are available for improving cognitive ability and maintaining a good quality of life; instituting such therapies early in the disease course is likely to have the greatest positive clinical effect.
Collapse
Affiliation(s)
- Curtis Wells Dewey
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, C4 169 Clinical Programs Center, Ithaca, NY 14853, USA.
| | - Emma S Davies
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, C4 169 Clinical Programs Center, Ithaca, NY 14853, USA
| | - Huisheng Xie
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA; Department of Comparative, Diagnostic and Population Medicine, 9700 Highway 318 West, Reddick, FL 32686, USA
| | - Joseph J Wakshlag
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA; Department of Comparative, Diagnostic and Population Medicine
| |
Collapse
|
249
|
Unsupervised excitation: GABAergic dysfunctions in Alzheimer’s disease. Brain Res 2019; 1707:216-226. [DOI: 10.1016/j.brainres.2018.11.042] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 10/31/2018] [Accepted: 11/27/2018] [Indexed: 12/22/2022]
|
250
|
Henstridge CM, Tzioras M, Paolicelli RC. Glial Contribution to Excitatory and Inhibitory Synapse Loss in Neurodegeneration. Front Cell Neurosci 2019; 13:63. [PMID: 30863284 PMCID: PMC6399113 DOI: 10.3389/fncel.2019.00063] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 02/08/2019] [Indexed: 12/12/2022] Open
Abstract
Synapse loss is an early feature shared by many neurodegenerative diseases, and it represents the major correlate of cognitive impairment. Recent studies reveal that microglia and astrocytes play a major role in synapse elimination, contributing to network dysfunction associated with neurodegeneration. Excitatory and inhibitory activity can be affected by glia-mediated synapse loss, resulting in imbalanced synaptic transmission and subsequent synaptic dysfunction. Here, we review the recent literature on the contribution of glia to excitatory/inhibitory imbalance, in the context of the most common neurodegenerative disorders. A better understanding of the mechanisms underlying pathological synapse loss will be instrumental to design targeted therapeutic interventions, taking in account the emerging roles of microglia and astrocytes in synapse remodeling.
Collapse
Affiliation(s)
- Christopher M Henstridge
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom.,Dementia Research Institute UK, The University of Edinburgh, Edinburgh, United Kingdom
| | - Makis Tzioras
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom.,Dementia Research Institute UK, The University of Edinburgh, Edinburgh, United Kingdom
| | - Rosa C Paolicelli
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|