1
|
Zhu WH, Yang XX, Gou XZ, Fu SM, Chen JH, Gao F, Shen Y, Bi DL, Tang AH. Nanoscale reorganisation of synaptic proteins in Alzheimer's disease. Neuropathol Appl Neurobiol 2023; 49:e12924. [PMID: 37461203 DOI: 10.1111/nan.12924] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 05/30/2023] [Accepted: 06/24/2023] [Indexed: 08/31/2023]
Abstract
AIMS Synaptic strength depends strongly on the subsynaptic organisation of presynaptic transmitter release and postsynaptic receptor densities, and their alterations are expected to underlie pathologies. Although synaptic dysfunctions are common pathogenic traits of Alzheimer's disease (AD), it remains unknown whether synaptic protein nano-organisation is altered in AD. Here, we systematically characterised the alterations in the subsynaptic organisation in cellular and mouse models of AD. METHODS We used immunostaining and super-resolution stochastic optical reconstruction microscopy imaging to quantitatively examine the synaptic protein nano-organisation in both Aβ1-42-treated neuronal cultures and cortical sections from a mouse model of AD, APP23 mice. RESULTS We found that Aβ1-42-treatment of cultured hippocampal neurons decreased the synaptic retention of postsynaptic scaffolds and receptors and disrupted their nanoscale alignment to presynaptic transmitter release sites. In cortical sections, we found that while GluA1 receptors in wild-type mice were organised in subsynaptic nanoclusters with high local densities, receptors in APP23 mice distributed more homogeneously within synapses. This reorganisation, together with the reduced overall receptor density, led to reduced glutamatergic synaptic transmission. Meanwhile, the transsynaptic alignment between presynaptic release-guiding RIM1/2 and postsynaptic scaffolding protein PSD-95 was reduced in APP23 mice. Importantly, these reorganisations were progressive with age and were more pronounced in synapses in close vicinity of Aβ plaques with dense cores. CONCLUSIONS Our study revealed a spatiotemporal-specific reorganisation of synaptic nanostructures in AD and identifies dense-core amyloid plaques as the major local inductor in APP23 mice.
Collapse
Affiliation(s)
- Wang-Hui Zhu
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Xiao-Xu Yang
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Xu-Zhuo Gou
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Shu-Mei Fu
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Jia-Hui Chen
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Feng Gao
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
| | - Yong Shen
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, China
| | - Dan-Lei Bi
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, China
| | - Ai-Hui Tang
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| |
Collapse
|
2
|
Lee AK, Yi N, Khaled H, Feller B, Takahashi H. SorCS1 inhibits amyloid-β binding to neurexin and rescues amyloid-β-induced synaptic pathology. Life Sci Alliance 2023; 6:e202201681. [PMID: 36697254 PMCID: PMC9880023 DOI: 10.26508/lsa.202201681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/27/2023] Open
Abstract
Amyloid-β oligomers (AβOs), toxic peptide aggregates found in Alzheimer's disease, cause synapse pathology. AβOs interact with neurexins (NRXs), key synaptic organizers, and this interaction dampens normal trafficking and function of NRXs. Axonal trafficking of NRX is in part regulated by its interaction with SorCS1, a protein sorting receptor, but the impact of SorCS1 regulation of NRXs in Aβ pathology was previously unstudied. Here, we show competition between the SorCS1 ectodomain and AβOs for β-NRX binding and rescue effects of the SorCS1b isoform on AβO-induced synaptic pathology. Like AβOs, the SorCS1 ectodomain binds to NRX1β through the histidine-rich domain of NRX1β, and the SorCS1 ectodomain and AβOs compete for NRX1β binding. In cultured hippocampal neurons, SorCS1b colocalizes with NRX1β on the axon surface, and axonal expression of SorCS1b rescues AβO-induced impairment of NRX-mediated presynaptic organization and presynaptic vesicle recycling and AβO-induced structural defects in excitatory synapses. Thus, our data suggest a role for SorCS1 in the rescue of AβO-induced NRX dysfunction and synaptic pathology, providing the basis for a novel potential therapeutic strategy for Alzheimer's disease.
Collapse
Affiliation(s)
- Alfred Kihoon Lee
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Canada
| | - Nayoung Yi
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Husam Khaled
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Benjamin Feller
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Hideto Takahashi
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
- Division of Experimental Medicine, McGill University, Montreal, Canada
| |
Collapse
|
3
|
Ribarič S. Detecting Early Cognitive Decline in Alzheimer's Disease with Brain Synaptic Structural and Functional Evaluation. Biomedicines 2023; 11:355. [PMID: 36830892 PMCID: PMC9952956 DOI: 10.3390/biomedicines11020355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Early cognitive decline in patients with Alzheimer's (AD) is associated with quantifiable structural and functional connectivity changes in the brain. AD dysregulation of Aβ and tau metabolism progressively disrupt normal synaptic function, leading to loss of synapses, decreased hippocampal synaptic density and early hippocampal atrophy. Advances in brain imaging techniques in living patients have enabled the transition from clinical signs and symptoms-based AD diagnosis to biomarkers-based diagnosis, with functional brain imaging techniques, quantitative EEG, and body fluids sampling. The hippocampus has a central role in semantic and episodic memory processing. This cognitive function is critically dependent on normal intrahippocampal connections and normal hippocampal functional connectivity with many cortical regions, including the perirhinal and the entorhinal cortex, parahippocampal cortex, association regions in the temporal and parietal lobes, and prefrontal cortex. Therefore, decreased hippocampal synaptic density is reflected in the altered functional connectivity of intrinsic brain networks (aka large-scale networks), including the parietal memory, default mode, and salience networks. This narrative review discusses recent critical issues related to detecting AD-associated early cognitive decline with brain synaptic structural and functional markers in high-risk or neuropsychologically diagnosed patients with subjective cognitive impairment or mild cognitive impairment.
Collapse
Affiliation(s)
- Samo Ribarič
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
4
|
Demuth H, Hosseini S, Düsedeau HP, Dunay IR, Korte M, Zagrebelsky M. Deletion of p75 NTR rescues the synaptic but not the inflammatory status in the brain of a mouse model for Alzheimer's disease. Front Mol Neurosci 2023; 16:1163087. [PMID: 37213691 PMCID: PMC10198655 DOI: 10.3389/fnmol.2023.1163087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/06/2023] [Indexed: 05/23/2023] Open
Abstract
Introduction Alzheimer's disease (AD), is characterized by a gradual cognitive decline associated with the accumulation of Amyloid beta (Aβ)-oligomers, progressive neuronal degeneration and chronic neuroinflammation. Among the receptors shown to bind and possibly transduce the toxic effects of Aβ-oligomers is the p75 neurotrophin receptor (p75NTR). Interestingly, p75NTR mediates several crucial processes in the nervous system, including neuronal survival and apoptosis, maintenance of the neuronal architecture, and plasticity. Furthermore, p75NTR is also expressed in microglia, the resident immune cells of the brain, where it is markedly increased under pathological conditions. These observations indicate p75NTR as a potential candidate for mediating Aβ-induced toxic effects at the interface between the nervous and the immune system, thereby potentially participating in the crosstalk between these two systems. Methods Here we used APP/PS1 transgenic mice (APP/PS1tg) and compared the Aβ-induced alterations in neuronal function, chronic inflammation as well as their cognitive consequences between 10 months old APP/PS1tg and APP/PS1tg x p75NTRexonIV knockout mice. Results Electrophysiological recordings show that a loss of p75NTR rescues the impairment in long-term potentiation at the Schaffer collaterals in the hippocampus of APP/PS1tg mice. Interestingly, however loss of p75NTR does not influence the severity of neuroinflammation, microglia activation or the decline in spatial learning and memory processes observed in APP/PS1tg mice. Conclusion Together these results indicate that while a deletion of p75NTR rescues the synaptic defect and the impairment in synaptic plasticity, it does not affect the progression of the neuroinflammation and the cognitive decline in a mouse model for AD.
Collapse
Affiliation(s)
- Hendrik Demuth
- Division of Cellular Neurobiology, Zoological Institute, Braunschweig, Germany
- Research Group Neuroinflammation and Neurodegeneration, Helmholtz Centre for Infection Research, AG NIND, Braunschweig, Germany
| | - Shirin Hosseini
- Division of Cellular Neurobiology, Zoological Institute, Braunschweig, Germany
- Research Group Neuroinflammation and Neurodegeneration, Helmholtz Centre for Infection Research, AG NIND, Braunschweig, Germany
| | - Henning Peter Düsedeau
- Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-von- Guericke University, Magdeburg, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-von- Guericke University, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, Braunschweig, Germany
- Research Group Neuroinflammation and Neurodegeneration, Helmholtz Centre for Infection Research, AG NIND, Braunschweig, Germany
| | - Marta Zagrebelsky
- Division of Cellular Neurobiology, Zoological Institute, Braunschweig, Germany
- *Correspondence: Marta Zagrebelsky,
| |
Collapse
|
5
|
Wu J, Su Y, Zhu W, Mallak NJ, Lepore N, Reiman EM, Caselli RJ, Thompson PM, Chen K, Wang Y. Improved Prediction of Amyloid-β and Tau Burden Using Hippocampal Surface Multivariate Morphometry Statistics and Sparse Coding. J Alzheimers Dis 2023; 91:637-651. [PMID: 36463452 PMCID: PMC9940990 DOI: 10.3233/jad-220812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Amyloid-β (Aβ) plaques and tau protein tangles in the brain are the defining 'A' and 'T' hallmarks of Alzheimer's disease (AD), and together with structural atrophy detectable on brain magnetic resonance imaging (MRI) scans as one of the neurodegenerative ('N') biomarkers comprise the "ATN framework" of AD. Current methods to detect Aβ/tau pathology include cerebrospinal fluid (invasive), positron emission tomography (PET; costly and not widely available), and blood-based biomarkers (promising but mainly still in development). OBJECTIVE To develop a non-invasive and widely available structural MRI-based framework to quantitatively predict the amyloid and tau measurements. METHODS With MRI-based hippocampal multivariate morphometry statistics (MMS) features, we apply our Patch Analysis-based Surface Correntropy-induced Sparse coding and max-pooling (PASCS-MP) method combined with the ridge regression model to individual amyloid/tau measure prediction. RESULTS We evaluate our framework on amyloid PET/MRI and tau PET/MRI datasets from the Alzheimer's Disease Neuroimaging Initiative. Each subject has one pair consisting of a PET image and MRI scan, collected at about the same time. Experimental results suggest that amyloid/tau measurements predicted with our PASCP-MP representations are closer to the real values than the measures derived from other approaches, such as hippocampal surface area, volume, and shape morphometry features based on spherical harmonics. CONCLUSION The MMS-based PASCP-MP is an efficient tool that can bridge hippocampal atrophy with amyloid and tau pathology and thus help assess disease burden, progression, and treatment effects.
Collapse
Affiliation(s)
- Jianfeng Wu
- School of Computing and Augmented Intelligence, Arizona State University, Tempe, AZ, USA
| | - Yi Su
- Banner Alzheimer’s Institute, Phoenix, AZ, USA
| | - Wenhui Zhu
- School of Computing and Augmented Intelligence, Arizona State University, Tempe, AZ, USA
| | - Negar Jalili Mallak
- School of Computing and Augmented Intelligence, Arizona State University, Tempe, AZ, USA
| | - Natasha Lepore
- CIBORG Lab, Department of Radiology Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | | | | | - Paul M. Thompson
- Imaging Genetics Center, Stevens Neuroimaging and Informatics Institute, University of Southern California, Marina del Rey, CA, USA
| | - Kewei Chen
- Banner Alzheimer’s Institute, Phoenix, AZ, USA
| | - Yalin Wang
- School of Computing and Augmented Intelligence, Arizona State University, Tempe, AZ, USA
| | | |
Collapse
|
6
|
Varshavskaya KB, Mitkevich VA, Makarov AA, Barykin EP. Synthetic, Cell-Derived, Brain-Derived, and Recombinant β-Amyloid: Modelling Alzheimer's Disease for Research and Drug Development. Int J Mol Sci 2022; 23:15036. [PMID: 36499362 PMCID: PMC9738609 DOI: 10.3390/ijms232315036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 12/02/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in the elderly, characterised by the accumulation of senile plaques and tau tangles, neurodegeneration, and neuroinflammation in the brain. The development of AD is a pathological cascade starting according to the amyloid hypothesis with the accumulation and aggregation of the β-amyloid peptide (Aβ), which induces hyperphosphorylation of tau and promotes the pro-inflammatory activation of microglia leading to synaptic loss and, ultimately, neuronal death. Modelling AD-related processes is important for both studying the molecular basis of the disease and the development of novel therapeutics. The replication of these processes is often achieved with the use of a purified Aβ peptide. However, Aβ preparations obtained from different sources can have strikingly different properties. This review aims to compare the structure and biological effects of Aβ oligomers and aggregates of a higher order: synthetic, recombinant, purified from cell culture, or extracted from brain tissue. The authors summarise the applicability of Aβ preparations for modelling Aβ aggregation, neurotoxicity, cytoskeleton damage, receptor toxicity in vitro and cerebral amyloidosis, synaptic plasticity disruption, and cognitive impairment in vivo and ex vivo. Further, the paper discusses the causes of the reported differences in the effect of Aβ obtained from the sources mentioned above. This review points to the importance of the source of Aβ for AD modelling and could help researchers to choose the optimal way to model the Aβ-induced abnormalities.
Collapse
Affiliation(s)
| | | | - Alexander A. Makarov
- Engelhardt Institute of Molecular Biology, Vavilov St. 32, 119991 Moscow, Russia
| | | |
Collapse
|
7
|
Piscopo P, Crestini A, Carbone E, Rivabene R, Ancidoni A, Lo Giudice M, Corbo M, Vanacore N, Lacorte E. A systematic review on drugs for synaptic plasticity in the treatment of dementia. Ageing Res Rev 2022; 81:101726. [PMID: 36031056 DOI: 10.1016/j.arr.2022.101726] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 01/31/2023]
Abstract
The aim of the present systematic review (SR) was to provide an overview of all published and unpublished clinical trials investigating the safety and efficacy of disease-modifying drugs targeting synaptic plasticity in dementia. Searches on CT.gov and EuCT identified 27 trials (4 phase-1, 1 phase-1/2, 18 phase-2, 1 phase-2/3, 1 phase-3, 1 phase-4, and 1 not reported). Twenty of them completed, and seven are currently active or enrolling. The structured bibliographic searches yielded 3585 records. A total of 12 studies were selected on Levetiracetam, Masitinib, Saracatinib, BI 40930, Bryostatin 1, PF-04447943 and Edonerpic drugs. We used RoB tool for quality analysis of randomized studies. Efficacy was assessed as a primary outcome in all studies except one and the main scale used was ADAS-Cog (7 studies), MMSE and CDR (4 studies). Safety and tolerability were reported in eleven studies. The incidence of SAEs was similar between treatment and placebo. At the moment, only one molecule reached phase-3. This could suggest that research on these drugs is still preliminary. Of all, three studies reported promising results on Levetiracetam, Bryostatin 1 and Masitinib.
Collapse
Affiliation(s)
- P Piscopo
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy.
| | - A Crestini
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - E Carbone
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - R Rivabene
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - A Ancidoni
- National Center for Disease Prevention ad Heath Promotion, Italian National Institute of Health, Rome, Italy
| | - M Lo Giudice
- Need Institute, Foundation for Cure and Rehabilitation of Neurological Diseases, Milan, Italy
| | - M Corbo
- Department of Neurorehabilitation Sciences, Casa Cura Policlinico, Milan, Italy.
| | - N Vanacore
- National Center for Disease Prevention ad Heath Promotion, Italian National Institute of Health, Rome, Italy
| | - E Lacorte
- National Center for Disease Prevention ad Heath Promotion, Italian National Institute of Health, Rome, Italy
| |
Collapse
|
8
|
Luchena C, Zuazo-Ibarra J, Valero J, Matute C, Alberdi E, Capetillo-Zarate E. A Neuron, Microglia, and Astrocyte Triple Co-culture Model to Study Alzheimer’s Disease. Front Aging Neurosci 2022; 14:844534. [PMID: 35493929 PMCID: PMC9048896 DOI: 10.3389/fnagi.2022.844534] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Glial cells are essential to understand Alzheimer’s disease (AD) progression, given their role in neuroinflammation and neurodegeneration. There is a need for reliable and easy to manipulate models that allow studying the mechanisms behind neuron and glia communication. Currently available models such as co-cultures require complex methodologies and/or might not be affordable for all laboratories. With this in mind, we aimed to establish a straightforward in vitro setting with neurons and glial cells to study AD. We generated and optimized a 2D triple co-culture model with murine astrocytes, neurons and microglia, based on sequential seeding of each cell type. Immunofluorescence, western blot and ELISA techniques were used to characterize the effects of oligomeric Aβ (oAβ) in this model. We found that, in the triple co-culture, microglia increased the expression of anti-inflammatory marker Arginase I, and reduced pro-inflammatory iNOS and IL-1β, compared with microglia alone. Astrocytes reduced expression of pro-inflammatory A1 markers AMIGO2 and C3, and displayed a ramified morphology resembling physiological conditions. Anti-inflammatory marker TGF-β1 was also increased in the triple co-culture. Lastly, neurons increased post-synaptic markers, and developed more and longer branches than in individual primary cultures. Addition of oAβ in the triple co-culture reduced synaptic markers and increased CD11b in microglia, which are hallmarks of AD. Consequently, we developed a straightforward and reproducible triple co-cultured model, where cells resemble physiological conditions better than in individual primary cultures: microglia are less inflammatory, astrocytes are less reactive and neurons display a more mature morphology. Moreover, we are able to recapitulate Aβ-induced synaptic loss and CD11b increase. This model emerges as a powerful tool to study neurodegeneration and neuroinflammation in the context of AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Celia Luchena
- Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- CIBERNED, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas, Madrid, Spain
| | - Jone Zuazo-Ibarra
- Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- CIBERNED, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas, Madrid, Spain
| | - Jorge Valero
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Institute of Neurosciences of Castilla y León, University of Salamanca, Salamanca, Spain
- Institute for Biomedical Research of Salamanca, Salamanca, Spain
| | - Carlos Matute
- Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- CIBERNED, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas, Madrid, Spain
| | - Elena Alberdi
- Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- CIBERNED, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas, Madrid, Spain
| | - Estibaliz Capetillo-Zarate
- Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- CIBERNED, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas, Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- *Correspondence: Estibaliz Capetillo-Zarate,
| |
Collapse
|
9
|
Wang Z, Zheng P, Xie Y, Chen X, Solowij N, Green K, Chew YL, Huang XF. Cannabidiol regulates CB1-pSTAT3 signaling for neurite outgrowth, prolongs lifespan, and improves health span in Caenorhabditis elegans of Aβ pathology models. FASEB J 2021; 35:e21537. [PMID: 33817834 DOI: 10.1096/fj.202002724r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/22/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022]
Abstract
Cannabidiol (CBD), a phytocannabinoid from the Cannabis sativa plant, exhibits a broad spectrum of potential therapeutic properties for neurodegenerative diseases. An accumulation of amyloid-β (Aβ) protein is one of the most important neuropathology in neurodegenerative diseases like Alzheimer's disease (AD). Data on the effect of CBD on the amelioration of Aβ-induced neurite degeneration and its consequences of life and health spans is sparse. This study aimed to investigate the effects of CBD on neurite outgrowth in cells and lifespan and health span in Caenorhabditis elegans (C. elegans). In human SH-SY5Y neuronal cells, CBD prevented neurite lesion induced by Aβ1-42 and increased the expression of fatty acid amide hydrolase (FAAH) and cannabinoid receptor 1 (CB1R). Furthermore, CBD both protected the reduction of dendritic spine density and rescued the activity of synaptic Ca2+ /calmodulin-dependent protein kinase II (CaMKII) from Aβ1-42 toxicity in primary hippocampal neurons. In C. elegans, we used the transgenic CL2355 strain of C. elegans, which expresses the human Aβ peptide throughout the nervous system and found that CBD treatment extended lifespan and improved health span. The neuroprotective effect of CBD was further explored by observing the dopaminergic neurons using transgenic dat-1: GFP strains using the confocal microscope. This study shows that CBD prevents the neurite degeneration induced by Aβ, by a mechanism involving CB1R activation, and extends lifespan and improves health span in Aβ-overexpressing worms. Our findings support the potential therapeutic approach of CBD for the treatment of AD patients.
Collapse
Affiliation(s)
- Zhizhen Wang
- Australian Centre for Cannabinoid Clinical and Research Excellence (ACRE), New Lambton Heights, NSW, Australia.,Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Peng Zheng
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Yuanyi Xie
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Xi Chen
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Nadia Solowij
- Australian Centre for Cannabinoid Clinical and Research Excellence (ACRE), New Lambton Heights, NSW, Australia.,Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia.,School of Psychology, University of Wollongong, Wollongong, NSW, Australia
| | - Katrina Green
- Australian Centre for Cannabinoid Clinical and Research Excellence (ACRE), New Lambton Heights, NSW, Australia.,Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia.,Molecular Horizons, School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Yee Lian Chew
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia.,Molecular Horizons, School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Xu-Feng Huang
- Australian Centre for Cannabinoid Clinical and Research Excellence (ACRE), New Lambton Heights, NSW, Australia.,Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia.,Molecular Horizons, School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| |
Collapse
|
10
|
Natesh SR, Hummels AR, Sachleben JR, Sosnick TR, Freed KF, Douglas JF, Meredith SC, Haddadian EJ. Molecular dynamics study of water channels in natural and synthetic amyloid-β fibrils. J Chem Phys 2021; 154:235102. [PMID: 34241272 PMCID: PMC8214467 DOI: 10.1063/5.0049250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/12/2021] [Indexed: 11/14/2022] Open
Abstract
We compared all-atom explicit solvent molecular dynamics simulations of three types of Aβ(1-40) fibrils: brain-seeded fibrils (2M4J, with a threefold axial symmetry) and the other two, all-synthetic fibril polymorphs (2LMN and 2LMP, made under different fibrillization conditions). Fibril models were constructed using either a finite or an infinite number of layers made using periodic images. These studies yielded four conclusions. First, finite fibrils tend to unravel in a manner reminiscent of fibril dissolution, while infinite fibrils were more stable during simulations. Second, salt bridges in these fibrils remained stable in those fibrils that contained them initially, and those without salt bridges did not develop them over the time course of the simulations. Third, all fibrils tended to develop a "stagger" or register shift of β-strands along the fibril axis. Fourth and most importantly, the brain-seeded, 2M4J, infinite fibrils allowed bidirectional transport of water in and out of the central longitudinal core of the fibril by rapidly developing gaps at the fibril vertices. 2LMP fibrils also showed this behavior, although to a lesser extent. The diffusion of water molecules in the fibril core region involved two dynamical states: a localized state and directed diffusion in the presence of obstacles. These observations provided support for the hypothesis that Aβ fibrils could act as nanotubes. At least some Aβ oligomers resembled fibrils structurally in having parallel, in-register β-sheets and a sheet-turn-sheet motif. Thus, our findings could have implications for Aβ cytotoxicity, which may occur through the ability of oligomers to form abnormal water and ion channels in cell membranes.
Collapse
Affiliation(s)
- S. R. Natesh
- Biological Sciences Collegiate Division, The University of Chicago, Chicago, Illinois 60637, USA
| | - A. R. Hummels
- Biological Sciences Collegiate Division, The University of Chicago, Chicago, Illinois 60637, USA
| | - J. R. Sachleben
- Division of Biological Sciences, The University of Chicago, Chicago, Illinois 60637, USA
| | - T. R. Sosnick
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, Illinois 60637, USA
| | - K. F. Freed
- James Franck Institute, The University of Chicago, Chicago, Illinois 60637, USA
| | - J. F. Douglas
- Material Measurement Laboratory, Material Science and Engineering Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, USA
| | - S. C. Meredith
- Departments of Pathology, Biochemistry, and Molecular Biology, The University of Chicago, Chicago, Illinois 60637, USA
| | - E. J. Haddadian
- Biological Sciences Collegiate Division, The University of Chicago, Chicago, Illinois 60637, USA
| |
Collapse
|
11
|
Mahaman YAR, Huang F, Embaye KS, Wang X, Zhu F. The Implication of STEP in Synaptic Plasticity and Cognitive Impairments in Alzheimer's Disease and Other Neurological Disorders. Front Cell Dev Biol 2021; 9:680118. [PMID: 34195199 PMCID: PMC8236946 DOI: 10.3389/fcell.2021.680118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/06/2021] [Indexed: 12/31/2022] Open
Abstract
STriatal-Enriched protein tyrosine Phosphatase (STEP) is a tyrosine phosphatase that has been implicated in Alzheimer’s disease (AD), the most common form of dementia, and many other neurological diseases. The protein level and activity of STEP have been found to be elevated in most of these disorders, and specifically in AD as a result of dysregulation of different pathways including PP2B/DARPP32/PP1, PKA as well as impairments of both proteasomal and lysosomal systems. The upregulation in STEP leads to increased binding to, and dephosphorylation of, its substrates which are mainly found to be synaptic plasticity and thus learning and memory related proteins. These proteins include kinases like Fyn, Pyk2, ERK1/2 and both NMDA and AMPA receptor subunits GluN2B and GluA2. The dephosphorylation of these molecules results in inactivation of these kinases and internalization of NMDA and AMPA receptor complexes leading to synapse loss and cognitive impairments. In this study, we aim to review STEP regulation and its implications in AD as well as other neurological disorders and then summarize data on targeting STEP as therapeutic strategy in these diseases.
Collapse
Affiliation(s)
- Yacoubou Abdoul Razak Mahaman
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital, Shenzhen University, Shenzhen, China.,Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Huang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kidane Siele Embaye
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaochuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Feiqi Zhu
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital, Shenzhen University, Shenzhen, China
| |
Collapse
|
12
|
Back MK, Ruggieri S, Jacobi E, von Engelhardt J. Amyloid Beta-Mediated Changes in Synaptic Function and Spine Number of Neocortical Neurons Depend on NMDA Receptors. Int J Mol Sci 2021; 22:ijms22126298. [PMID: 34208315 PMCID: PMC8231237 DOI: 10.3390/ijms22126298] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 11/16/2022] Open
Abstract
Onset and progression of Alzheimer's disease (AD) pathophysiology differs between brain regions. The neocortex, for example, is a brain region that is affected very early during AD. NMDA receptors (NMDARs) are involved in mediating amyloid beta (Aβ) toxicity. NMDAR expression, on the other hand, can be affected by Aβ. We tested whether the high vulnerability of neocortical neurons for Aβ-toxicity may result from specific NMDAR expression profiles or from a particular regulation of NMDAR expression by Aβ. Electrophysiological analyses suggested that pyramidal cells of 6-months-old wildtype mice express mostly GluN1/GluN2A NMDARs. While synaptic NMDAR-mediated currents are unaltered in 5xFAD mice, extrasynaptic NMDARs seem to contain GluN1/GluN2A and GluN1/GluN2A/GluN2B. We used conditional GluN1 and GluN2B knockout mice to investigate whether NMDARs contribute to Aβ-toxicity. Spine number was decreased in pyramidal cells of 5xFAD mice and increased in neurons with 3-week virus-mediated Aβ-overexpression. NMDARs were required for both Aβ-mediated changes in spine number and functional synapses. Thus, our study gives novel insights into the Aβ-mediated regulation of NMDAR expression and the role of NMDARs in Aβ pathophysiology in the somatosensory cortex.
Collapse
|
13
|
Caspase Activation and Caspase-Mediated Cleavage of APP Is Associated with Amyloid β-Protein-Induced Synapse Loss in Alzheimer's Disease. Cell Rep 2021; 31:107839. [PMID: 32610140 DOI: 10.1016/j.celrep.2020.107839] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 01/29/2020] [Accepted: 06/08/2020] [Indexed: 01/09/2023] Open
Abstract
Amyloid β-protein (Aβ) toxicity is hypothesized to play a seminal role in Alzheimer's disease (AD) pathogenesis. However, it remains unclear how Aβ causes synaptic dysfunction and synapse loss. We hypothesize that one mechanism of Aβ-induced synaptic injury is related to the cleavage of amyloid β precursor protein (APP) at position D664 by caspases that release the putatively cytotoxic C31 peptide. In organotypic slice cultures derived from mice with a knock-in mutation in the APP gene (APP D664A) to inhibit caspase cleavage, Aβ-induced synaptic injury is markedly reduced in two models of Aβ toxicity. Loss of dendritic spines is also attenuated in mice treated with caspase inhibitors. Importantly, the time-dependent dendritic spine loss is correlated with localized activation of caspase-3 but is absent in APP D664A cultures. We propose that the APP cytosolic domain plays an essential role in Aβ-induced synaptic damage in the injury pathway mediated by localized caspase activation.
Collapse
|
14
|
Izzo NJ, Yuede CM, LaBarbera KM, Limegrover CS, Rehak C, Yurko R, Waybright L, Look G, Rishton G, Safferstein H, Hamby ME, Williams C, Sadlek K, Edwards HM, Davis CS, Grundman M, Schneider LS, DeKosky ST, Chelsky D, Pike I, Henstridge C, Blennow K, Zetterberg H, LeVine H, Spires-Jones TL, Cirrito JR, Catalano SM. Preclinical and clinical biomarker studies of CT1812: A novel approach to Alzheimer's disease modification. Alzheimers Dement 2021; 17:1365-1382. [PMID: 33559354 PMCID: PMC8349378 DOI: 10.1002/alz.12302] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/16/2020] [Accepted: 01/02/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Amyloid beta (Aβ) oligomers are one of the most toxic structural forms of the Aβ protein and are hypothesized to cause synaptotoxicity and memory failure as they build up in Alzheimer's disease (AD) patients' brain tissue. We previously demonstrated that antagonists of the sigma-2 receptor complex effectively block Aβ oligomer toxicity. CT1812 is an orally bioavailable, brain penetrant small molecule antagonist of the sigma-2 receptor complex that appears safe and well tolerated in healthy elderly volunteers. We tested CT1812's effect on Aβ oligomer pathobiology in preclinical AD models and evaluated CT1812's impact on cerebrospinal fluid (CSF) protein biomarkers in mild to moderate AD patients in a clinical trial (ClinicalTrials.gov NCT02907567). METHODS Experiments were performed to measure the impact of CT1812 versus vehicle on Aβ oligomer binding to synapses in vitro, to human AD patient post mortem brain tissue ex vivo, and in living APPSwe /PS1dE9 transgenic mice in vivo. Additional experiments were performed to measure the impact of CT1812 versus vehicle on Aβ oligomer-induced deficits in membrane trafficking rate, synapse number, and protein expression in mature hippocampal/cortical neurons in vitro. The impact of CT1812 on cognitive function was measured in transgenic Thy1 huAPPSwe/Lnd+ and wild-type littermates. A multicenter, double-blind, placebo-controlled parallel group trial was performed to evaluate the safety, tolerability, and impact on protein biomarker expression of CT1812 or placebo given once daily for 28 days to AD patients (Mini-Mental State Examination 18-26). CSF protein expression was measured by liquid chromatography with tandem mass spectrometry or enzyme-linked immunosorbent assay in samples drawn prior to dosing (Day 0) and at end of dosing (Day 28) and compared within each patient and between pooled treated versus placebo-treated dosing groups. RESULTS CT1812 significantly and dose-dependently displaced Aβ oligomers bound to synaptic receptors in three independent preclinical models of AD, facilitated oligomer clearance into the CSF, increased synaptic number and protein expression in neurons, and improved cognitive performance in transgenic mice. CT1812 significantly increased CSF concentrations of Aβ oligomers in AD patient CSF, reduced concentrations of synaptic proteins and phosphorylated tau fragments, and reversed expression of many AD-related proteins dysregulated in CSF. DISCUSSION These preclinical studies demonstrate the novel disease-modifying mechanism of action of CT1812 against AD and Aβ oligomers. The clinical results are consistent with preclinical data and provide evidence of target engagement and impact on fundamental disease-related signaling pathways in AD patients, supporting further development of CT1812.
Collapse
Affiliation(s)
| | | | | | | | - Courtney Rehak
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, USA
| | - Raymond Yurko
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, USA
| | - Lora Waybright
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, USA
| | - Gary Look
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, USA
| | | | | | - Mary E Hamby
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, USA
| | | | - Kelsey Sadlek
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, USA
| | | | | | - Michael Grundman
- Global R&D Partners, San Diego, California, USA.,University of California San Diego, San Diego, California, USA
| | - Lon S Schneider
- Keck School of Medicine of USC, Los Angeles, California, USA
| | - Steven T DeKosky
- McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | | | | | | | - Kaj Blennow
- University of Gothenburg, Mölndal, Sweden.,Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- University of Gothenburg, Mölndal, Sweden.,Sahlgrenska University Hospital, Mölndal, Sweden.,UCL Institute of Neurology, London, UK
| | - Harry LeVine
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | | | | | | |
Collapse
|
15
|
Chatterjee M, Kwon J, Benedict J, Kamceva M, Kurup P, Lombroso PJ. STEP inhibition prevents Aβ-mediated damage in dendritic complexity and spine density in Alzheimer's disease. Exp Brain Res 2021; 239:881-890. [PMID: 33420799 DOI: 10.1007/s00221-020-06028-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 12/28/2020] [Indexed: 11/30/2022]
Abstract
Loss of dendritic spines and decline of cognitive function are hallmarks of patients with Alzheimer's disease (AD). Previous studies have shown that AD pathophysiology involves increased expression of a central nervous system-enriched protein tyrosine phosphatase called STEP (STriatal-Enriched protein tyrosine Phosphatase). STEP opposes the development of synaptic strengthening by dephosphorylating substrates, including GluN2B, Pyk2, and ERK1/2. Genetic reduction of STEP as well as pharmacological inhibition of STEP improve cognitive function and hippocampal memory in the 3×Tg-AD mouse model. Here, we show that the improved cognitive function is accompanied by an increase in synaptic connectivity in cell cultures as well as in the triple transgenic AD mouse model, further highlighting the potential of STEP inhibitors as a therapeutic agent.
Collapse
Affiliation(s)
- Manavi Chatterjee
- Child Study Center, Yale University, 230 South Frontage Rd, New Haven, CT, 06520, USA.
| | - Jeemin Kwon
- Child Study Center, Yale University, 230 South Frontage Rd, New Haven, CT, 06520, USA
| | - Jessie Benedict
- Child Study Center, Yale University, 230 South Frontage Rd, New Haven, CT, 06520, USA
| | - Marija Kamceva
- Child Study Center, Yale University, 230 South Frontage Rd, New Haven, CT, 06520, USA
| | - Pradeep Kurup
- Child Study Center, Yale University, 230 South Frontage Rd, New Haven, CT, 06520, USA.,Department of Surgery, University of Alabama at Birmingham, 1900 University Blvd, Birmingham, AL, 35233, United States
| | - Paul J Lombroso
- Child Study Center, Yale University, 230 South Frontage Rd, New Haven, CT, 06520, USA. .,Departments of Psychiatry, Yale University, New Haven, CT, 06520, USA. .,Departments of Neurobiology, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
16
|
Ca 2+ Dyshomeostasis Disrupts Neuronal and Synaptic Function in Alzheimer's Disease. Cells 2020; 9:cells9122655. [PMID: 33321866 PMCID: PMC7763805 DOI: 10.3390/cells9122655] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
Ca2+ homeostasis is essential for multiple neuronal functions and thus, Ca2+ dyshomeostasis can lead to widespread impairment of cellular and synaptic signaling, subsequently contributing to dementia and Alzheimer's disease (AD). While numerous studies implicate Ca2+ mishandling in AD, the cellular basis for loss of cognitive function remains under investigation. The process of synaptic degradation and degeneration in AD is slow, and constitutes a series of maladaptive processes each contributing to a further destabilization of the Ca2+ homeostatic machinery. Ca2+ homeostasis involves precise maintenance of cytosolic Ca2+ levels, despite extracellular influx via multiple synaptic Ca2+ channels, and intracellular release via organelles such as the endoplasmic reticulum (ER) via ryanodine receptor (RyRs) and IP3R, lysosomes via transient receptor potential mucolipin channel (TRPML) and two pore channel (TPC), and mitochondria via the permeability transition pore (PTP). Furthermore, functioning of these organelles relies upon regulated inter-organelle Ca2+ handling, with aberrant signaling resulting in synaptic dysfunction, protein mishandling, oxidative stress and defective bioenergetics, among other consequences consistent with AD. With few effective treatments currently available to mitigate AD, the past few years have seen a significant increase in the study of synaptic and cellular mechanisms as drivers of AD, including Ca2+ dyshomeostasis. Here, we detail some key findings and discuss implications for future AD treatments.
Collapse
|
17
|
Nakamura T, Oh CK, Liao L, Zhang X, Lopez KM, Gibbs D, Deal AK, Scott HR, Spencer B, Masliah E, Rissman RA, Yates JR, Lipton SA. Noncanonical transnitrosylation network contributes to synapse loss in Alzheimer's disease. Science 2020; 371:science.aaw0843. [PMID: 33273062 DOI: 10.1126/science.aaw0843] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 11/18/2020] [Indexed: 12/21/2022]
Abstract
Here we describe mechanistically distinct enzymes (a kinase, a guanosine triphosphatase, and a ubiquitin protein hydrolase) that function in disparate biochemical pathways and can also act in concert to mediate a series of redox reactions. Each enzyme manifests a second, noncanonical function-transnitrosylation-that triggers a pathological biochemical cascade in mouse models and in humans with Alzheimer's disease (AD). The resulting series of transnitrosylation reactions contributes to synapse loss, the major pathological correlate to cognitive decline in AD. We conclude that enzymes with distinct primary reaction mechanisms can form a completely separate network for aberrant transnitrosylation. This network operates in the postreproductive period, so natural selection against such abnormal activity may be decreased.
Collapse
Affiliation(s)
- Tomohiro Nakamura
- Departments of Molecular Medicine and Neuroscience, and Neuroscience Translational Center, The Scripps Research Institute, La Jolla, CA 92037, USA. .,Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA 92121, USA
| | - Chang-Ki Oh
- Departments of Molecular Medicine and Neuroscience, and Neuroscience Translational Center, The Scripps Research Institute, La Jolla, CA 92037, USA.,Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA 92121, USA
| | - Lujian Liao
- Departments of Molecular Medicine and Neuroscience, and Neuroscience Translational Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Xu Zhang
- Departments of Molecular Medicine and Neuroscience, and Neuroscience Translational Center, The Scripps Research Institute, La Jolla, CA 92037, USA.,Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA 92121, USA
| | - Kevin M Lopez
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA 92121, USA
| | - Daniel Gibbs
- Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Amanda K Deal
- Departments of Molecular Medicine and Neuroscience, and Neuroscience Translational Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Henry R Scott
- Departments of Molecular Medicine and Neuroscience, and Neuroscience Translational Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Brian Spencer
- Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA.,VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - John R Yates
- Departments of Molecular Medicine and Neuroscience, and Neuroscience Translational Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Stuart A Lipton
- Departments of Molecular Medicine and Neuroscience, and Neuroscience Translational Center, The Scripps Research Institute, La Jolla, CA 92037, USA. .,Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA 92121, USA.,Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| |
Collapse
|
18
|
Patnaik A, Zagrebelsky M, Korte M, Holz A. Signaling via the p75 neurotrophin receptor facilitates amyloid-β-induced dendritic spine pathology. Sci Rep 2020; 10:13322. [PMID: 32770070 PMCID: PMC7415136 DOI: 10.1038/s41598-020-70153-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 07/20/2020] [Indexed: 12/21/2022] Open
Abstract
Synapse and dendritic spine loss induced by amyloid-β oligomers is one of the main hallmarks of the early phases of Alzheimer's disease (AD) and is directly correlated with the cognitive decline typical of this pathology. The p75 neurotrophin receptor (p75NTR) binds amyloid-β oligomers in the nM range. While it was shown that µM concentrations of amyloid-β mediate cell death, the role and intracellular signaling of p75NTR for dendritic spine pathology induced by sublethal concentrations of amyloid-β has not been analyzed. We describe here p75NTR as a crucial binding partner in mediating effects of soluble amyloid-β oligomers on dendritic spine density and structure in non-apoptotic hippocampal neurons. Removing or over-expressing p75NTR in neurons rescues or exacerbates the typical loss of dendritic spines and their structural alterations observed upon treatment with nM concentrations of amyloid-β oligomers. Moreover, we show that binding of amyloid-β oligomers to p75NTR activates the RhoA/ROCK signaling cascade resulting in the fast stabilization of the actin spinoskeleton. Our results describe a role for p75NTR and downstream signaling events triggered by binding of amyloid-β oligomers and causing dendritic spine pathology. These observations further our understanding of the molecular mechanisms underlying one of the main early neuropathological hallmarks of AD.
Collapse
Affiliation(s)
- Abhisarika Patnaik
- Zoological Institute, Division of Cellular Neurobiology, TU Braunschweig, 38108, Braunschweig, Germany
| | - Marta Zagrebelsky
- Zoological Institute, Division of Cellular Neurobiology, TU Braunschweig, 38108, Braunschweig, Germany
| | - Martin Korte
- Zoological Institute, Division of Cellular Neurobiology, TU Braunschweig, 38108, Braunschweig, Germany
- Group Neuroinflammation and Neurodegeneration, Helmholtz Centre for Infection Research, AG NIND, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Andreas Holz
- Group Neuroinflammation and Neurodegeneration, Helmholtz Centre for Infection Research, AG NIND, Inhoffenstrasse 7, 38124, Braunschweig, Germany.
| |
Collapse
|
19
|
Nguyen TT, Ta QTH, Nguyen TKO, Nguyen TTD, Vo VG. Role of Body-Fluid Biomarkers in Alzheimer's Disease Diagnosis. Diagnostics (Basel) 2020; 10:diagnostics10050326. [PMID: 32443860 PMCID: PMC7277970 DOI: 10.3390/diagnostics10050326] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/02/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD) is a complex neurodegenerative disease that requires extremely specific biomarkers for its diagnosis. For current diagnostics capable of identifying AD, the development and validation of early stage biomarkers is a top research priority. Body-fluid biomarkers might closely reflect synaptic dysfunction in the brain and, thereby, could contribute to improving diagnostic accuracy and monitoring disease progression, and serve as markers for assessing the response to disease-modifying therapies at early onset. Here, we highlight current advances in the research on the capabilities of body-fluid biomarkers and their role in AD pathology. Then, we describe and discuss current applications of the potential biomarkers in clinical diagnostics in AD.
Collapse
Affiliation(s)
- Thuy Trang Nguyen
- Faculty of Pharmacy, Ho Chi Minh City University of Technology (HUTECH), Ho Chi Minh City 700000, Vietnam;
| | - Qui Thanh Hoai Ta
- Institute of Research and Development, Duy Tan University, Danang 550000, Vietnam;
| | - Thi Kim Oanh Nguyen
- Faculty of Food Science and Technology, Ho Chi Minh City University of Food Industry, Ho Chi Minh City 700000, Vietnam;
| | - Thi Thuy Dung Nguyen
- Faculty of Environmental and Food Engineering, Nguyen Tat Thanh University, Ho Chi Minh City 70000, Vietnam
- Correspondence: (T.T.D.N.); (V.G.V.)
| | - Van Giau Vo
- Department of Industrial and Environmental Engineering, Graduate School of Environment, Gachon University, 1342 Sungnam-daero, Sujung-gu, Seongnam-si, Gyeonggi-do 461-701, Korea
- Department of BionanoTechnology, Gachon University, 1342 Sungnam-daero, Sujung-gu, Seongnam-si, Gyeonggi-do 461-701, Korea
- Correspondence: (T.T.D.N.); (V.G.V.)
| |
Collapse
|
20
|
Crystal JD. Memory: Amyloid Beta Is Good Before It Is Bad. Curr Biol 2020; 30:R449-R450. [PMID: 32428478 DOI: 10.1016/j.cub.2020.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A fundamental mystery in the biology of memory is to understand the pathway from normal memory to later dysfunctional memory. Some insight on this problem comes from new research suggesting that amyloid beta helps memory consolidation, before it impairs memory.
Collapse
Affiliation(s)
- Jonathon D Crystal
- Department of Psychological and Brain Sciences, Indiana University, 1101 E. 10th Street, Bloomington, IN 47405-7007, USA.
| |
Collapse
|
21
|
Colom-Cadena M, Spires-Jones T, Zetterberg H, Blennow K, Caggiano A, DeKosky ST, Fillit H, Harrison JE, Schneider LS, Scheltens P, de Haan W, Grundman M, van Dyck CH, Izzo NJ, Catalano SM. The clinical promise of biomarkers of synapse damage or loss in Alzheimer's disease. Alzheimers Res Ther 2020; 12:21. [PMID: 32122400 PMCID: PMC7053087 DOI: 10.1186/s13195-020-00588-4] [Citation(s) in RCA: 168] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/14/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND Synapse damage and loss are fundamental to the pathophysiology of Alzheimer's disease (AD) and lead to reduced cognitive function. The goal of this review is to address the challenges of forging new clinical development approaches for AD therapeutics that can demonstrate reduction of synapse damage or loss. The key points of this review include the following: Synapse loss is a downstream effect of amyloidosis, tauopathy, inflammation, and other mechanisms occurring in AD.Synapse loss correlates most strongly with cognitive decline in AD because synaptic function underlies cognitive performance.Compounds that halt or reduce synapse damage or loss have a strong rationale as treatments of AD.Biomarkers that measure synapse degeneration or loss in patients will facilitate clinical development of such drugs.The ability of methods to sensitively measure synapse density in the brain of a living patient through synaptic vesicle glycoprotein 2A (SV2A) positron emission tomography (PET) imaging, concentrations of synaptic proteins (e.g., neurogranin or synaptotagmin) in the cerebrospinal fluid (CSF), or functional imaging techniques such as quantitative electroencephalography (qEEG) provides a compelling case to use these types of measurements as biomarkers that quantify synapse damage or loss in clinical trials in AD. CONCLUSION A number of emerging biomarkers are able to measure synapse injury and loss in the brain and may correlate with cognitive function in AD. These biomarkers hold promise both for use in diagnostics and in the measurement of therapeutic successes.
Collapse
Affiliation(s)
- Martí Colom-Cadena
- Centre for Discovery Brain Sciences, UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
| | - Tara Spires-Jones
- Centre for Discovery Brain Sciences, UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | | | - Steven T DeKosky
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Howard Fillit
- Alzheimer's Drug Discovery Foundation, New York, NY, USA
| | - John E Harrison
- Metis Cognition Ltd, Kilmington, UK
- Alzheimer Center, AUmc, Amsterdam, The Netherlands
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | | | - Phillip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Willem de Haan
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Clinical Neurophysiology and MEG, VU University Medical Center, Amsterdam, Netherlands
| | | | - Christopher H van Dyck
- Alzheimer's Disease Research Unit and Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | | | | |
Collapse
|
22
|
Gutierrez DA, Vargas LM, Chandia-Cristi A, de la Fuente C, Leal N, Alvarez AR. c-Abl Deficiency Provides Synaptic Resiliency Against Aβ-Oligomers. Front Cell Neurosci 2019; 13:526. [PMID: 31849613 PMCID: PMC6902026 DOI: 10.3389/fncel.2019.00526] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022] Open
Abstract
Spine pathology has been implicated in the early onset of Alzheimer’s disease (AD), where Aβ-Oligomers (AβOs) cause synaptic dysfunction and loss. Previously, we described that pharmacological inhibition of c-Abl prevents AβOs-induced synaptic alterations. Hence, this kinase seems to be a key element in AD progression. Here, we studied the role of c-Abl on dendritic spine morphological changes induced by AβOs using c-Abl null neurons (c-Abl-KO). First, we characterized the effect of c-Abl deficiency on dendritic spine density and found that its absence increases dendritic spine density. While AβOs-treatment reduces the spine number in both wild-type (WT) and c-Abl-KO neurons, AβOs-driven spine density loss was not affected by c-Abl. We then characterized AβOs-induced morphological changes in dendritic spines of c-Abl-KO neurons. AβOs induced a decrease in the number of mushroom spines in c-Abl-KO neurons while preserving the populations of immature stubby, thin, and filopodia spines. Furthermore, synaptic contacts evaluated by PSD95/Piccolo clustering and cell viability were preserved in AβOs-exposed c-Abl-KO neurons. In conclusion, our results indicate that in the presence of AβOs c-Abl participates in synaptic contact removal, increasing susceptibility to AβOs damage. Its deficiency increases the immature spine population reducing AβOs-induced synapse elimination. Therefore, c-Abl signaling could be a relevant actor in the early stages of AD.
Collapse
Affiliation(s)
- Daniela A Gutierrez
- Cell Signaling Laboratory, Faculty of Biological Science, Department of Cell and Molecular Biology, Center for Aging and Regeneration (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Lina M Vargas
- Cell Signaling Laboratory, Faculty of Biological Science, Department of Cell and Molecular Biology, Center for Aging and Regeneration (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - América Chandia-Cristi
- Cell Signaling Laboratory, Faculty of Biological Science, Department of Cell and Molecular Biology, Center for Aging and Regeneration (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina de la Fuente
- Cell Signaling Laboratory, Faculty of Biological Science, Department of Cell and Molecular Biology, Center for Aging and Regeneration (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nancy Leal
- Cell Signaling Laboratory, Faculty of Biological Science, Department of Cell and Molecular Biology, Center for Aging and Regeneration (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandra R Alvarez
- Cell Signaling Laboratory, Faculty of Biological Science, Department of Cell and Molecular Biology, Center for Aging and Regeneration (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
23
|
Mehra R, Kepp KP. Cell size effects in the molecular dynamics of the intrinsically disordered Aβ peptide. J Chem Phys 2019; 151:085101. [DOI: 10.1063/1.5115085] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Rukmankesh Mehra
- Technical University of Denmark, DTU Chemistry, Building 206, 2800 Kgs. Lyngby, Denmark
| | - Kasper P. Kepp
- Technical University of Denmark, DTU Chemistry, Building 206, 2800 Kgs. Lyngby, Denmark
| |
Collapse
|
24
|
Shi Y, Fang YY, Wei YP, Jiang Q, Zeng P, Tang N, Lu Y, Tian Q. Melatonin in Synaptic Impairments of Alzheimer's Disease. J Alzheimers Dis 2019; 63:911-926. [PMID: 29710712 DOI: 10.3233/jad-171178] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) underlies dementia for millions of people worldwide with no effective treatment. The dementia of AD is thought stem from the impairments of the synapses because of their critical roles in cognition. Melatonin is a neurohormone mainly released by the pineal gland in a circadian manner and it regulates brain functions in various manners. It is reported that both the melatonin deficit and synaptic impairments are present in the very early stage of AD and strongly contribute to the progress of AD. In the mammalian brains, the effects of melatonin are mainly relayed by two of its receptors, melatonin receptor type 1a (MT1) and 1b (MT2). To have a clear idea on the roles of melatonin in synaptic impairments of AD, this review discussed the actions of melatonin and its receptors in the stabilization of synapses, modulation of long-term potentiation, as well as their contributions in the transmissions of glutamatergic, GABAergic and dopaminergic synapses, which are the three main types of synapses relevant to the synaptic strength. The synaptic protective roles of melatonin in AD treatment were also summarized. Regarding its protective roles against amyloid-β neurotoxicity, tau hyperphosphorylation, oxygenation, inflammation as well as synaptic dysfunctions, melatonin may be an ideal therapeutic agent against AD at early stage.
Collapse
Affiliation(s)
- Yan Shi
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, China
| | - Ying-Yan Fang
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Ping Wei
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Jiang
- Integrated TCM and Western Medicine Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Zeng
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, China
| | - Na Tang
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, China
| | - Youming Lu
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Tian
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
25
|
Xie Z, Shapiro LP, Cahill ME, Russell TA, Lacor PN, Klein WL, Penzes P. Kalirin-7 prevents dendritic spine dysgenesis induced by amyloid beta-derived oligomers. Eur J Neurosci 2019; 49:1091-1101. [PMID: 30565792 DOI: 10.1111/ejn.14311] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/19/2018] [Accepted: 12/13/2018] [Indexed: 12/15/2022]
Abstract
Synapse degeneration and dendritic spine dysgenesis are believed to be crucial early steps in Alzheimer's disease (AD), and correlate with cognitive deficits in AD patients. Soluble amyloid beta (Aβ)-derived oligomers, also termed Aβ-derived diffusible ligands (ADDLs), accumulate in the brain of AD patients and play a crucial role in AD pathogenesis. ADDLs bind to mature hippocampal neurons, induce structural changes in dendritic spines and contribute to neuronal death. However, mechanisms underlying structural and toxic effects are not fully understood. Here, we report that ADDLs bind to cultured mature cortical pyramidal neurons and induce spine dysgenesis. ADDL treatment induced the rapid depletion of kalirin-7, a brain-specific guanine-nucleotide exchange factor for the small GTPase Rac1, from spines. Kalirin-7 is a key regulator of dendritic spine morphogenesis and maintenance in forebrain pyramidal neurons and here we show that overexpression of kalirin-7 prevents ADDL-induced spine degeneration. Taken together, our results suggest that kalirin-7 may play a role in the early events leading to synapse degeneration, and its pharmacological activation may prevent or delay synapse pathology in AD.
Collapse
Affiliation(s)
- Zhong Xie
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Lauren P Shapiro
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Michael E Cahill
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Theron A Russell
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Pascale N Lacor
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, Illinois
| | - William L Klein
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, Illinois
| | - Peter Penzes
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
26
|
Belikov AV. Age-related diseases as vicious cycles. Ageing Res Rev 2019; 49:11-26. [PMID: 30458244 DOI: 10.1016/j.arr.2018.11.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 10/05/2018] [Accepted: 11/15/2018] [Indexed: 02/07/2023]
Abstract
The mortality rates of age-related diseases (ARDs) increase exponentially with age. Processes described by the exponential growth function typically involve a branching chain reaction or, more generally, a positive feedback loop. Here I propose that each ARD is mediated by one or several positive feedback loops (vicious cycles). I then identify critical vicious cycles in five major ARDs: atherosclerosis, hypertension, diabetes, Alzheimer's and Parkinson's. I also propose that the progression of ARDs can be halted by selectively interrupting the vicious cycles and suggest the most promising targets.
Collapse
Affiliation(s)
- Aleksey V Belikov
- Laboratory of Innovative Medicine, School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Institutsky per., 9, 141701 Dolgoprudny, Moscow Region, Russia.
| |
Collapse
|
27
|
Müller MK, Jacobi E, Sakimura K, Malinow R, von Engelhardt J. NMDA receptors mediate synaptic depression, but not spine loss in the dentate gyrus of adult amyloid Beta (Aβ) overexpressing mice. Acta Neuropathol Commun 2018; 6:110. [PMID: 30352630 PMCID: PMC6198500 DOI: 10.1186/s40478-018-0611-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 09/29/2018] [Indexed: 11/25/2022] Open
Abstract
Amyloid beta (Aβ)-mediated synapse dysfunction and spine loss are considered to be early events in Alzheimer’s disease (AD) pathogenesis. N-methyl-D-aspartate receptors (NMDARs) have previously been suggested to play a role for Amyloid beta (Aβ) toxicity. Pharmacological block of NMDAR subunits in cultured neurons and mice suggested that NMDARs containing the GluN2B subunit are necessary for Aβ-mediated changes in synapse number and function in hippocampal neurons. Interestingly, NMDARs undergo a developmental switch from GluN2B- to GluN2A-containing receptors. This indicates different functional roles of NMDARs in young mice compared to older animals. In addition, the lack of pharmacological tools to efficiently dissect the role of NMDARs containing the different subunits complicates the interpretation of their specific role. In order to address this problem and to investigate the specific role for Aβ toxicity of the distinct NMDAR subunits in dentate gyrus granule cells of adult mice, we used conditional knockout mouse lines for the subunits GluN1, GluN2A and GluN2B. Aβ-mediated changes in synaptic function and neuronal anatomy were investigated in several-months old mice with virus-mediated overproduction of Aβ and in 1-year old 5xFAD mice. We found that all three NMDAR subunits contribute to the Aβ-mediated decrease in the number of functional synapses. However, NMDARs are not required for the spine number reduction in dentate gyrus granule cells after chronic Aβ-overproduction in 5xFAD mice. Furthermore, the amplitude of synaptic and extrasynaptic NMDAR-mediated currents was reduced in dentate gyrus granule of 5xFAD mice without changes in current kinetics, suggesting that a redistribution or change in subunit composition of NMDARs does not play a role in mediating Amyloid beta (Aβ) toxicity. Our study indicates that NMDARs are involved in AD pathogenesis by compromising synapse function but not by affecting neuron morphology.
Collapse
|
28
|
Lim SL, Tran DN, Zumkehr J, Chen C, Ghiaar S, Kieu Z, Villanueva E, Gallup V, Rodriguez-Ortiz CJ, Kitazawa M. Inhibition of hematopoietic cell kinase dysregulates microglial function and accelerates early stage Alzheimer's disease-like neuropathology. Glia 2018; 66:2700-2718. [PMID: 30277607 DOI: 10.1002/glia.23522] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 08/02/2018] [Accepted: 08/02/2018] [Indexed: 11/08/2022]
Abstract
Emerging evidence have posited that dysregulated microglia impair clearance and containment of amyloid-β (Aβ) species in the brain, resulting in aberrant buildup of Aβ and onset of Alzheimer's disease (AD). Hematopoietic cell kinase (Hck) is one of the key regulators of phagocytosis among the Src family tyrosine kinases (SFKs) in myeloid cells, and its expression is found to be significantly altered in AD brains. However, the role of Hck signaling in AD pathogenesis is unknown. We employed pharmacological inhibition and genetic ablation of Hck in BV2 microglial cells and J20 mouse model of AD, respectively, to evaluate the impact of Hck deficiency on Aβ-stimulated microglial phagocytosis, Aβ clearance, and resultant AD-like neuropathology. Our in vitro data reveal that pharmacological inhibition of SFKs/Hck in BV2 cells and genetic ablation of their downstream kinase, spleen tyrosine kinase (Syk), in primary microglia significantly attenuate Aβ oligomers-stimulated microglial phagocytosis. Whereas in Hck-deficient J20 mice, we observed exacerbated Aβ plaque burden, reduced microglial coverage, containment, and phagocytosis of Aβ plaques, and induced iNOS expression in plaque-associated microglial clusters. These multifactorial changes in microglial activities led to attenuated PSD95 levels in hippocampal DG and CA3 regions, but did not alter the postsynaptic dendritic spine morphology at the CA1 region nor cognitive function of the mice. Hck inhibition thus accelerates early stage AD-like neuropathology by dysregulating microglial function and inducing neuroinflammation. Our data implicate that Hck pathway plays a prominent role in regulating microglial neuroprotective function during the early stage of AD development.
Collapse
Affiliation(s)
- Siok Lam Lim
- Center for Occupational and Environmental Health, Department of Medicine, University of California, Irvine, California.,Molecular and Cell Biology, University of California, Merced, California
| | - Diana Nguyen Tran
- Molecular and Cell Biology, University of California, Merced, California
| | - Joannee Zumkehr
- Center for Occupational and Environmental Health, Department of Medicine, University of California, Irvine, California.,Molecular and Cell Biology, University of California, Merced, California
| | - Christine Chen
- Molecular and Cell Biology, University of California, Merced, California
| | - Sagar Ghiaar
- Molecular and Cell Biology, University of California, Merced, California
| | - Zanett Kieu
- Molecular and Cell Biology, University of California, Merced, California
| | | | - Victoria Gallup
- Molecular and Cell Biology, University of California, Merced, California
| | - Carlos J Rodriguez-Ortiz
- Center for Occupational and Environmental Health, Department of Medicine, University of California, Irvine, California.,Molecular and Cell Biology, University of California, Merced, California
| | - Masashi Kitazawa
- Center for Occupational and Environmental Health, Department of Medicine, University of California, Irvine, California.,Molecular and Cell Biology, University of California, Merced, California
| |
Collapse
|
29
|
Chiang ACA, Fowler SW, Savjani RR, Hilsenbeck SG, Wallace CE, Cirrito JR, Das P, Jankowsky JL. Combination anti-Aβ treatment maximizes cognitive recovery and rebalances mTOR signaling in APP mice. J Exp Med 2018; 215:1349-1364. [PMID: 29626114 PMCID: PMC5940263 DOI: 10.1084/jem.20171484] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 01/03/2018] [Accepted: 03/07/2018] [Indexed: 01/01/2023] Open
Abstract
Chiang et al. show that combining two complementary approaches for Aβ reduction improved cognitive function in a mouse model of amyloidosis relative to either treatment alone. Efficacy corresponded with restoration of mTOR signaling, TFEB expression, and autophagic flux, suggesting additional targets for future polytherapy in AD. Drug development for Alzheimer’s disease has endeavored to lower amyloid β (Aβ) by either blocking production or promoting clearance. The benefit of combining these approaches has been examined in mouse models and shown to improve pathological measures of disease over single treatment; however, the impact on cellular and cognitive functions affected by Aβ has not been tested. We used a controllable APP transgenic mouse model to test whether combining genetic suppression of Aβ production with passive anti-Aβ immunization improved functional outcomes over either treatment alone. Compared with behavior before treatment, arresting further Aβ production (but not passive immunization) was sufficient to stop further decline in spatial learning, working memory, and associative memory, whereas combination treatment reversed each of these impairments. Cognitive improvement coincided with resolution of neuritic dystrophy, restoration of synaptic density surrounding deposits, and reduction of hyperactive mammalian target of rapamycin signaling. Computational modeling corroborated by in vivo microdialysis pointed to the reduction of soluble/exchangeable Aβ as the primary driver of cognitive recovery.
Collapse
Affiliation(s)
- Angie C A Chiang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX
| | | | | | - Susan G Hilsenbeck
- Department of Medicine, Lester and Sue Smith Breast Center, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Clare E Wallace
- Department of Neurology, Knight Alzheimer's Disease Research Center, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO
| | - John R Cirrito
- Department of Neurology, Knight Alzheimer's Disease Research Center, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO
| | - Pritam Das
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL
| | - Joanna L Jankowsky
- Department of Neuroscience, Baylor College of Medicine, Houston, TX .,Departments of Neurology, Neurosurgery, and Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| |
Collapse
|
30
|
Park D, Chang S. Soluble Aβ 1-42 increases the heterogeneity in synaptic vesicle pool size among synapses by suppressing intersynaptic vesicle sharing. Mol Brain 2018; 11:10. [PMID: 29463281 PMCID: PMC5819658 DOI: 10.1186/s13041-018-0353-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 02/13/2018] [Indexed: 11/10/2022] Open
Abstract
Growing evidence has indicated that prefibrillar form of soluble amyloid beta (sAβ1-42) is the major causative factor in the synaptic dysfunction associated with AD. The molecular changes leading to presynaptic dysfunction caused by sAβ1-42, however, still remains elusive. Recently, we found that sAβ1-42 inhibits chemically induced long-term potentiation-induced synaptogenesis by suppressing the intersynaptic vesicle trafficking through calcium (Ca2+) dependent hyperphosphorylation of synapsin and CaMKIV. However, it is still unclear how sAβ1-42 increases intracellular Ca2+ that induces hyperphosphorylation of CaMKIV and synapsin, and what is the functional consequences of sAβ1-42-induced defects in intersynaptic vesicle trafficking in physiological conditions. In this study, we showed that sAβ1-42elevated intracellular Ca2+ through not only extracellular Ca2+ influx but also Ca2+ release from mitochondria. Surprisingly, without Ca2+ release from mitochondria, sAβ1-42 failed to increase intracellular Ca2+ even in the presence of normal extracellular Ca2+. We further found that sAβ1-42-induced mitochondria Ca2+ release alone sufficiently increased Serine 9 phosphorylation of synapsin. By blocking synaptic vesicle reallocation, sAβ1-42 significantly increased heterogeneity of total synaptic vesicle pool size among synapses. Together, our results suggested that by disrupting the axonal vesicle trafficking, sAβ1-42 disabled neurons to adjust synaptic pool sizes among synapses, which might prevent homeostatic rescaling in synaptic strength of individual neurons.
Collapse
Affiliation(s)
- Daehun Park
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Sunghoe Chang
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea. .,Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, 03080, South Korea.
| |
Collapse
|
31
|
Reduced expression of Na +/Ca 2+ exchangers is associated with cognitive deficits seen in Alzheimer's disease model mice. Neuropharmacology 2017; 131:291-303. [PMID: 29274751 DOI: 10.1016/j.neuropharm.2017.12.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 12/08/2017] [Accepted: 12/20/2017] [Indexed: 11/21/2022]
Abstract
Na+/Ca2+ exchangers (NCXs) are expressed primarily in the plasma membrane of most cell types, where they mediate electrogenic exchange of one Ca2+ for three Na+ ions, depending on Ca2+ and Na+ electrochemical gradients across the membrane. Three mammalian NCX isoforms (NCX1, NCX2, and NCX3) are each encoded by a distinct gene. Here, we report that NCX2 and NCX3 protein and mRNA levels are relatively reduced in hippocampal CA1 of APP23 and APP-KI mice. Likewise, NCX2+/- or NCX3+/- mice exhibited impaired hippocampal LTP and memory-related behaviors. Moreover, relative to controls, calcium/calmodulin-dependent protein kinase II (CaMKII) autophosphorylation significantly decreased in NCX2+/- mouse hippocampus but increased in hippocampus of NCX3+/- mice. NCX2 or NCX3 heterozygotes displayed impaired maintenance of hippocampal LTP, a phenotype that in NCX2+/- mice was correlated with elevated calcineurin activity and rescued by treatment with the calcineurin (CaN) inhibitor FK506. Likewise, FK506 treatment significantly restored impaired hippocampal LTP in APP-KI mice. Moreover, Ca2+ clearance after depolarization following high frequency stimulation was slightly delayed in hippocampal CA1 regions of NCX2+/- mice. Electron microscopy revealed relatively decreased synaptic density in CA1 of NCX2+/- mice, while the number of spines with perforated synapses in CA1 significantly increased in NCX3+/- mice. We conclude that memory impairment seen in NCX2+/- and NCX3+/- mice reflect dysregulated hippocampal CaMKII activity, which alters dendritic spine morphology, findings with implications for memory deficits seen in Alzheimer's disease model mice.
Collapse
|
32
|
|
33
|
Park D, Na M, Kim JA, Lee U, Cho E, Jang M, Chang S. Activation of CaMKIV by soluble amyloid-β 1-42 impedes trafficking of axonal vesicles and impairs activity-dependent synaptogenesis. Sci Signal 2017; 10:10/487/eaam8661. [PMID: 28698220 DOI: 10.1126/scisignal.aam8661] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The prefibrillar form of soluble amyloid-β (sAβ1-42) impairs synaptic function and is associated with the early phase of Alzheimer's disease (AD). We investigated how sAβ1-42 led to presynaptic defects using a quantum dot-based, single particle-tracking method to monitor synaptic vesicle (SV) trafficking along axons. We found that sAβ1-42 prevented new synapse formation induced by chemical long-term potentiation (cLTP). In cultured rat hippocampal neurons, nanomolar amounts of sAβ1-42 impaired Ca2+ clearance from presynaptic terminals and increased the basal Ca2+ concentration. This caused an increase in the phosphorylation of Ca2+/calmodulin-dependent protein kinase IV (CaMKIV) and its substrate synapsin, which markedly inhibited SV trafficking along axons between synapses. Neurons derived from a transgenic AD mouse model had similar defects, which were prevented by an inhibitor of CaMK kinase (CaMKK; which activates CaMKIV), by antibodies against Aβ1-42, or by expression a phosphodeficient synapsin mutant. The CaMKK inhibitor also abolished the defects in activity-dependent synaptogenesis caused by sAβ1-42 Our results suggest that by disrupting SV reallocation between synapses, sAβ1-42 prevents neurons from forming new synapses or adjusting strength and activity among neighboring synapses. Targeting this mechanism might prevent synaptic dysfunction in AD patients.
Collapse
Affiliation(s)
- Daehun Park
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Myeongsu Na
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Jung Ah Kim
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, South Korea.,Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Unghwi Lee
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Eunji Cho
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Mirye Jang
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, South Korea.,Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Sunghoe Chang
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, South Korea. .,Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul 110-799, South Korea.,Biomembrane Plasticity Research Center, Seoul National University College of Medicine, Seoul 110-799, South Korea
| |
Collapse
|
34
|
Kocahan S, Doğan Z. Mechanisms of Alzheimer's Disease Pathogenesis and Prevention: The Brain, Neural Pathology, N-methyl-D-aspartate Receptors, Tau Protein and Other Risk Factors. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2017; 15:1-8. [PMID: 28138104 PMCID: PMC5290713 DOI: 10.9758/cpn.2017.15.1.1] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 06/28/2016] [Accepted: 07/07/2016] [Indexed: 12/31/2022]
Abstract
The characteristic features of Alzheimer’s disease (AD) are the appearance of extracellular amyloid-beta (Aβ) plaques and neurofibrillary tangles in the intracellular environment, neuronal death and the loss of synapses, all of which contribute to cognitive decline in a progressive manner. A number of hypotheses have been advanced to explain AD. Abnormal tau phosphorylation may contribute to the formation of abnormal neurofibrillary structures. Many different structures are susceptible to AD, including the reticular formation, the nuclei in the brain stem (e.g., raphe nucleus), thalamus, hypothalamus, locus ceruleus, amygdala, substantia nigra, striatum, and claustrum. Excitotoxicity results from continuous, low-level activation of N-methyl-D-aspartate (NMDA) receptors. Premature synaptotoxicity, changes in neurotransmitter expression, neurophils loss, accumulation of amyloid β-protein deposits (amyloid/senile plaques), and neuronal loss and brain atrophy are all associated with stages of AD progression. Several recent studies have examined the relationship between Aβ and NMDA receptors. Aβ-induced spine loss is associated with a decrease in glutamate receptors and is dependent upon the calcium-dependent phosphatase calcineurin, which has also been linked to long-term depression.
Collapse
Affiliation(s)
- Sayad Kocahan
- Department of Physiology, Faculty of Medicine, Adiyaman University, Adiyaman, Turkey.,International Scientific Center, Baku State University, Baku, Azerbaijan
| | - Zumrut Doğan
- Department of Anatomy, Faculty of Medicine, Adiyaman University, Adiyaman, Turkey
| |
Collapse
|
35
|
Amyloid-β Oligomers Interact with Neurexin and Diminish Neurexin-mediated Excitatory Presynaptic Organization. Sci Rep 2017; 7:42548. [PMID: 28211900 PMCID: PMC5304201 DOI: 10.1038/srep42548] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/12/2017] [Indexed: 01/29/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by excessive production and deposition of amyloid-beta (Aβ) proteins as well as synapse dysfunction and loss. While soluble Aβ oligomers (AβOs) have deleterious effects on synapse function and reduce synapse number, the underlying molecular mechanisms are not well understood. Here we screened synaptic organizer proteins for cell-surface interaction with AβOs and identified a novel interaction between neurexins (NRXs) and AβOs. AβOs bind to NRXs via the N-terminal histidine-rich domain (HRD) of β-NRX1/2/3 and alternatively-spliced inserts at splicing site 4 of NRX1/2. In artificial synapse-formation assays, AβOs diminish excitatory presynaptic differentiation induced by NRX-interacting proteins including neuroligin1/2 (NLG1/2) and the leucine-rich repeat transmembrane protein LRRTM2. Although AβOs do not interfere with the binding of NRX1β to NLG1 or LRRTM2, time-lapse imaging revealed that AβO treatment reduces surface expression of NRX1β on axons and that this reduction depends on the NRX1β HRD. In transgenic mice expressing mutated human amyloid precursor protein, synaptic expression of β-NRXs, but not α-NRXs, decreases. Thus our data indicate that AβOs interact with NRXs and that this interaction inhibits NRX-mediated presynaptic differentiation by reducing surface expression of axonal β-NRXs, providing molecular and mechanistic insights into how AβOs lead to synaptic pathology in AD.
Collapse
|
36
|
Dore K, Aow J, Malinow R. The Emergence of NMDA Receptor Metabotropic Function: Insights from Imaging. Front Synaptic Neurosci 2016; 8:20. [PMID: 27516738 PMCID: PMC4963461 DOI: 10.3389/fnsyn.2016.00020] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 07/06/2016] [Indexed: 01/19/2023] Open
Abstract
The NMDA receptor (R) participates in many important physiological and pathological processes. For example, its activation is required for both long-term potentiation (LTP) and long-term depression (LTD) of synaptic transmission, cellular models of learning and memory. Furthermore, it may play a role in the actions of amyloid-beta on synapses as well as in the signaling leading to cell death following stroke. Until recently, these processes were thought to be mediated by ion-flux through the receptor. Using a combination of imaging and electrophysiological approaches, ion-flux independent functions of the NMDAR were recently examined. In this review, we will discuss the role of metabotropic NMDAR function in LTD and synaptic dysfunction.
Collapse
Affiliation(s)
- Kim Dore
- Center for Neural Circuits and Behavior, Department of Neuroscience and Section for Neurobiology, Division of Biology, University of California at San Diego San Diego, CA, USA
| | - Jonathan Aow
- Genome Institute of Singapore Singapore, Singapore
| | - Roberto Malinow
- Center for Neural Circuits and Behavior, Department of Neuroscience and Section for Neurobiology, Division of Biology, University of California at San Diego San Diego, CA, USA
| |
Collapse
|
37
|
Kepp KP. Alzheimer's disease due to loss of function: A new synthesis of the available data. Prog Neurobiol 2016; 143:36-60. [PMID: 27327400 DOI: 10.1016/j.pneurobio.2016.06.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/10/2016] [Accepted: 06/11/2016] [Indexed: 12/11/2022]
Abstract
Alzheimer's Disease (AD) is a highly complex disease involving a broad range of clinical, cellular, and biochemical manifestations that are currently not understood in combination. This has led to many views of AD, e.g. the amyloid, tau, presenilin, oxidative stress, and metal hypotheses. The amyloid hypothesis has dominated the field with its assumption that buildup of pathogenic β-amyloid (Aβ) peptide causes disease. This paradigm has been criticized, yet most data suggest that Aβ plays a key role in the disease. Here, a new loss-of-function hypothesis is synthesized that accounts for the anomalies of the amyloid hypothesis, e.g. the curious pathogenicity of the Aβ42/Aβ40 ratio, the loss of Aβ caused by presenilin mutation, the mixed phenotypes of APP mutations, the poor clinical-biochemical correlations for genetic variant carriers, and the failure of Aβ reducing drugs. The amyloid-loss view accounts for recent findings on the structure and chemical features of Aβ variants and their coupling to human patient data. The lost normal function of APP/Aβ is argued to be metal transport across neuronal membranes, a view with no apparent anomalies and substantially more explanatory power than the gain-of-function amyloid hypothesis. In the loss-of-function scenario, the central event of Aβ aggregation is interpreted as a loss of soluble, functional monomer Aβ rather than toxic overload of oligomers. Accordingly, new research models and treatment strategies should focus on remediation of the functional amyloid balance, rather than strict containment of Aβ, which, for reasons rationalized in this review, has failed clinically.
Collapse
Affiliation(s)
- Kasper P Kepp
- Technical University of Denmark, DTU Chemistry, DK-2800 Kongens Lyngby, Denmark.
| |
Collapse
|
38
|
Regulation of the Postsynaptic Compartment of Excitatory Synapses by the Actin Cytoskeleton in Health and Its Disruption in Disease. Neural Plast 2016; 2016:2371970. [PMID: 27127658 PMCID: PMC4835652 DOI: 10.1155/2016/2371970] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 03/09/2016] [Indexed: 02/07/2023] Open
Abstract
Disruption of synaptic function at excitatory synapses is one of the earliest pathological changes seen in wide range of neurological diseases. The proper control of the segregation of neurotransmitter receptors at these synapses is directly correlated with the intact regulation of the postsynaptic cytoskeleton. In this review, we are discussing key factors that regulate the structure and dynamics of the actin cytoskeleton, the major cytoskeletal building block that supports the postsynaptic compartment. Special attention is given to the complex interplay of actin-associated proteins that are found in the synaptic specialization. We then discuss our current understanding of how disruption of these cytoskeletal elements may contribute to the pathological events observed in the nervous system under disease conditions with a particular focus on Alzheimer's disease pathology.
Collapse
|
39
|
Luo HB, Li Y, Liu ZJ, Cao L, Zhang ZQ, Wang Y, Zhang XY, Liu Z, Shi XQ. Protective effect of tetrahydroxy stilbene glucoside on learning and memory by regulating synaptic plasticity. Neural Regen Res 2016; 11:1480-1486. [PMID: 27857754 PMCID: PMC5090853 DOI: 10.4103/1673-5374.191223] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Damage to synaptic plasticity induced by neurotoxicity of amyloid-beta is regarded to be one of the pathological mechanisms of learning and memory disabilities in Alzheimer's disease patients. This study assumed that the damage of amyloid-beta to learning and memory abilities was strongly associated with the changes in the Fyn/N-methyl-D-aspartate receptor 2B (NR2B) expression. An APP695V7171 transgenic mouse model of Alzheimer's disease was used and treatment with tetrahydroxy-stilbene glucoside was administered intragastrically. Results showed that intragastric administration of tetrahydroxy-stilbene glucoside improved the learning and memory abilities of the transgenic mice through increasing NR2B receptors and Fyn expression. It also reversed parameters for synaptic interface structure of gray type I. These findings indicate that tetrahydroxy stilbene glucoside has protective effects on the brain, and has prospects for its clinical application to improve the learning and memory abilities and treat Alzheimer's disease.
Collapse
Affiliation(s)
- Hong-Bo Luo
- Department of Neurology, Lanzhou General Hospital, Lanzhou Military Area Command, Lanzhou, Gansu Province, China
| | - Yun Li
- Department of Neurology, Lanzhou General Hospital, Lanzhou Military Area Command, Lanzhou, Gansu Province, China
| | - Zun-Jing Liu
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Li Cao
- Department of Neurology, Lanzhou General Hospital, Lanzhou Military Area Command, Lanzhou, Gansu Province, China
| | - Zhi-Qiang Zhang
- Department of Neurology, Lanzhou General Hospital, Lanzhou Military Area Command, Lanzhou, Gansu Province, China
| | - Yong Wang
- Department of Neurology, Lanzhou General Hospital, Lanzhou Military Area Command, Lanzhou, Gansu Province, China
| | - Xiao-Yan Zhang
- Department of Neurology, Lanzhou General Hospital, Lanzhou Military Area Command, Lanzhou, Gansu Province, China
| | - Zhao Liu
- Department of Neurology, Lanzhou General Hospital, Lanzhou Military Area Command, Lanzhou, Gansu Province, China
| | - Xiang-Qun Shi
- Department of Neurology, Lanzhou General Hospital, Lanzhou Military Area Command, Lanzhou, Gansu Province, China
| |
Collapse
|
40
|
Fernández-Fernández D, Dorner-Ciossek C, Kroker KS, Rosenbrock H. Age-related synaptic dysfunction in Tg2576 mice starts as a failure in early long-term potentiation which develops into a full abolishment of late long-term potentiation. J Neurosci Res 2015; 94:266-81. [PMID: 26629777 DOI: 10.1002/jnr.23701] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 09/30/2015] [Accepted: 11/17/2015] [Indexed: 11/12/2022]
Abstract
Tg2576 mice are widely used to study amyloid-dependent synaptic dysfunction related to Alzheimer's disease. However, conflicting data have been reported for these mice with regard to basal transmission as well as the in vitro correlate of memory, long-term potentiation (LTP). Some studies show clear impairments, whereas others report no deficiency. The present study uses hippocampal slices from 3-, 10-, and 15-month-old wild-type (WT) and Tg2576 mice to evaluate synaptic function in each group, including experiments to investigate basal synaptic transmission, short- and long-term plasticity by inducing paired-pulse facilitation, and both early and late LTP. We show that synaptic function remains intact in hippocampal slices from Tg2576 mice at 3 months of age. However, both early and late LTP decline progressively during aging in these mice. This deterioration of synaptic plasticity starts affecting early LTP, ultimately leading to the abolishment of both forms of LTP in 15-month-old animals. In comparison, WT littermates display normal synaptic parameters during aging. Additional pharmacological investigation into the involvement of NMDA receptors and L-type voltage-gated calcium channels in LTP suggests a distinct mechanism of induction among age groups, demonstrating that both early and late LTP are differentially affected by these channels in Tg2576 mice during aging.
Collapse
Affiliation(s)
- Diego Fernández-Fernández
- Deparment of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| | - Cornelia Dorner-Ciossek
- Deparment of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| | - Katja S Kroker
- Deptartment of Drug Discovery Support, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| | - Holger Rosenbrock
- Deparment of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| |
Collapse
|
41
|
Maiti P, Manna J, Ilavazhagan G, Rossignol J, Dunbar GL. Molecular regulation of dendritic spine dynamics and their potential impact on synaptic plasticity and neurological diseases. Neurosci Biobehav Rev 2015; 59:208-37. [PMID: 26562682 DOI: 10.1016/j.neubiorev.2015.09.020] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 08/20/2015] [Accepted: 09/07/2015] [Indexed: 12/12/2022]
Abstract
The structure and dynamics of dendritic spines reflect the strength of synapses, which are severely affected in different brain diseases. Therefore, understanding the ultra-structure, molecular signaling mechanism(s) regulating dendritic spine dynamics is crucial. Although, since last century, dynamics of spine have been explored by several investigators in different neurological diseases, but despite countless efforts, a comprehensive understanding of the fundamental etiology and molecular signaling pathways involved in spine pathology is lacking. The purpose of this review is to provide a contextual framework of our current understanding of the molecular mechanisms of dendritic spine signaling, as well as their potential impact on different neurodegenerative and psychiatric diseases, as a format for highlighting some commonalities in function, as well as providing a format for new insights and perspectives into this critical area of research. Additionally, the potential strategies to restore spine structure-function in different diseases are also pointed out. Overall, these informations should help researchers to design new drugs to restore the structure-function of dendritic spine, a "hot site" of synaptic plasticity.
Collapse
Affiliation(s)
- Panchanan Maiti
- Field Neurosciences Institute, St. Mary's of Michigan, Saginaw, MI, USA; Department of Psychology and Neurosciences Program, Central Michigan University, Mt. Pleasant, MI, USA.
| | - Jayeeta Manna
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA.
| | - G Ilavazhagan
- Hindustan University, Rajiv Gandhi Salai (OMR), Padur, Kelambakam, Chennai, TN, India.
| | - Julien Rossignol
- Department of Psychology and Neurosciences Program, Central Michigan University, Mt. Pleasant, MI, USA; College of Medicine, Central Michigan University, Mt. Pleasant, MI, USA.
| | - Gary L Dunbar
- Field Neurosciences Institute, St. Mary's of Michigan, Saginaw, MI, USA; Department of Psychology and Neurosciences Program, Central Michigan University, Mt. Pleasant, MI, USA.
| |
Collapse
|
42
|
Tong JQ, Zhang J, Hao M, Yang J, Han YF, Liu XJ, Shi H, Wu MN, Liu QS, Qi JS. Leptin attenuates the detrimental effects of β-amyloid on spatial memory and hippocampal later-phase long term potentiation in rats. Horm Behav 2015; 73:125-30. [PMID: 26135065 DOI: 10.1016/j.yhbeh.2015.06.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 04/21/2015] [Accepted: 06/25/2015] [Indexed: 12/26/2022]
Abstract
β-Amyloid (Aβ) is the main component of amyloid plaques developed in the brain of patients with Alzheimer's disease (AD). The increasing burden of Aβ in the cortex and hippocampus is closely correlated with memory loss and cognition deficits in AD. Recently, leptin, a 16kD peptide derived mainly from white adipocyte tissue, has been appreciated for its neuroprotective function, although less is known about the effects of leptin on spatial memory and synaptic plasticity. The present study investigated the neuroprotective effects of leptin against Aβ-induced deficits in spatial memory and in vivo hippocampal late-phase long-term potentiation (L-LTP) in rats. Y maze spontaneous alternation was used to assess short term working memory, and the Morris water maze task was used to assess long term reference memory. Hippocampal field potential recordings were performed to observe changes in L-LTP. We found that chronically intracerebroventricular injection of leptin (1μg) effectively alleviated Aβ1-42 (20μg)-induced spatial memory impairments of Y maze spontaneous alternation and Morris water maze. In addition, chronic administration of leptin also reversed Aβ1-42-induced suppression of in vivo hippocampal L-LTP in rats. Together, these results suggest that chronic leptin treatments reversed Aβ-induced deficits in learning and memory and the maintenance of L-LTP.
Collapse
Affiliation(s)
- Jia-Qing Tong
- Department of Neurobiology and National Key Discipline of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China; Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jun Zhang
- Department of Neurobiology and National Key Discipline of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Ming Hao
- Department of Neurobiology and National Key Discipline of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Ju Yang
- Department of Neurobiology and National Key Discipline of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Yu-Fei Han
- Department of Neurobiology and National Key Discipline of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Xiao-Jie Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Hui Shi
- Department of Neurobiology and National Key Discipline of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Mei-Na Wu
- Department of Neurobiology and National Key Discipline of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jin-Shun Qi
- Department of Neurobiology and National Key Discipline of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China.
| |
Collapse
|
43
|
Sevlever D, Zou F, Ma L, Carrasquillo S, Crump MG, Culley OJ, Hunter TA, Bisceglio GD, Younkin L, Allen M, Carrasquillo MM, Sando SB, Aasly JO, Dickson DW, Graff-Radford NR, Petersen RC, Deák F, Belbin O. Genetically-controlled Vesicle-Associated Membrane Protein 1 expression may contribute to Alzheimer's pathophysiology and susceptibility. Mol Neurodegener 2015; 10:18. [PMID: 25881291 PMCID: PMC4426163 DOI: 10.1186/s13024-015-0015-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 03/26/2015] [Indexed: 12/20/2022] Open
Abstract
Background Alzheimer’s disease is a neurodegenerative disorder in which extracellular deposition of β-amyloid (Aβ) oligomers causes synaptic injury resulting in early memory loss, altered homeostasis, accumulation of hyperphosphorylated tau and cell death. Since proteins in the SNAP (Soluble N-ethylmaleimide-sensitive factor Attachment Protein) REceptors (SNARE) complex are essential for neuronal Aβ release at pre-synaptic terminals, we hypothesized that genetically controlled SNARE expression could alter neuronal Aß release at the synapse and hence play an early role in Alzheimer’s pathophysiology. Results Here we report 5 polymorphisms in Vesicle-Associated Membrane Protein 1 (VAMP1), a gene encoding a member of the SNARE complex, associated with bidirectionally altered cerebellar VAMP1 transcript levels (all p < 0.05). At the functional level, we demonstrated that control of VAMP1 expression by heterogeneous knockdown in mice resulted in up to 74% reduction in neuronal Aβ exocytosis (p < 0.001). We performed a case-control association study of the 5 VAMP1 expression regulating polymorphisms in 4,667 Alzheimer’s disease patients and 6,175 controls to determine their contribution to Alzheimer’s disease risk. We found that polymorphisms associated with increased brain VAMP1 transcript levels conferred higher risk for Alzheimer’s disease than those associated with lower VAMP1 transcript levels (p = 0.03). Moreover, we also report a modest protective association for a common VAMP1 polymorphism with Alzheimer’s disease risk (OR = 0.88, p = 0.03). This polymorphism was associated with decreased VAMP1 transcript levels (p = 0.02) and was functionally active in a dual luciferase reporter gene assay (p < 0.01). Conclusions Genetically regulated VAMP1 expression in the brain may modify both Alzheimer’s disease risk and may contribute to Alzheimer’s pathophysiology. Electronic supplementary material The online version of this article (doi:10.1186/s13024-015-0015-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daniel Sevlever
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Fl, 32224, USA.
| | - Fanggeng Zou
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Fl, 32224, USA. .,Human Genetics Division, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA.
| | - Li Ma
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Fl, 32224, USA.
| | - Sebastian Carrasquillo
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Fl, 32224, USA.
| | - Michael G Crump
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Fl, 32224, USA.
| | - Oliver J Culley
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Fl, 32224, USA.
| | - Talisha A Hunter
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Fl, 32224, USA.
| | - Gina D Bisceglio
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Fl, 32224, USA.
| | - Linda Younkin
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Fl, 32224, USA.
| | - Mariet Allen
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Fl, 32224, USA.
| | - Minerva M Carrasquillo
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Fl, 32224, USA.
| | - Sigrid B Sando
- Department of Neurology, St. Olav's Hospital, Edvard Griegs Gate 8, 7006, Trondheim, Norway. .,Department of Neuroscience, Norwegian University of Science and Technology, NTNU, 7491, Trondheim, Norway.
| | - Jan O Aasly
- Department of Neuroscience, Norwegian University of Science and Technology, NTNU, 7491, Trondheim, Norway.
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Fl, 32224, USA.
| | - Neill R Graff-Radford
- Department of Neurology, Mayo Clinic College of Medicine, Jacksonville, Fl, 32224, USA.
| | - Ronald C Petersen
- Department of Neurology and the Mayo Alzheimer Disease Research Center, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
| | | | | | - Olivia Belbin
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Fl, 32224, USA. .,Memory Disorders Unit, Institute of Biomedical Investigation Sant Pau (IIB Sant Pau), Autonomous University of Barcelona (UAB), Barcelona, Spain.
| |
Collapse
|
44
|
Izzo NJ, Staniszewski A, To L, Fa M, Teich AF, Saeed F, Wostein H, Walko T, Vaswani A, Wardius M, Syed Z, Ravenscroft J, Mozzoni K, Silky C, Rehak C, Yurko R, Finn P, Look G, Rishton G, Safferstein H, Miller M, Johanson C, Stopa E, Windisch M, Hutter-Paier B, Shamloo M, Arancio O, LeVine H, Catalano SM. Alzheimer's therapeutics targeting amyloid beta 1-42 oligomers I: Abeta 42 oligomer binding to specific neuronal receptors is displaced by drug candidates that improve cognitive deficits. PLoS One 2014; 9:e111898. [PMID: 25390368 PMCID: PMC4229098 DOI: 10.1371/journal.pone.0111898] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 10/02/2014] [Indexed: 01/09/2023] Open
Abstract
Synaptic dysfunction and loss caused by age-dependent accumulation of synaptotoxic beta amyloid (Abeta) 1-42 oligomers is proposed to underlie cognitive decline in Alzheimer's disease (AD). Alterations in membrane trafficking induced by Abeta oligomers mediates reduction in neuronal surface receptor expression that is the basis for inhibition of electrophysiological measures of synaptic plasticity and thus learning and memory. We have utilized phenotypic screens in mature, in vitro cultures of rat brain cells to identify small molecules which block or prevent the binding and effects of Abeta oligomers. Synthetic Abeta oligomers bind saturably to a single site on neuronal synapses and induce deficits in membrane trafficking in neuronal cultures with an EC50 that corresponds to its binding affinity. The therapeutic lead compounds we have found are pharmacological antagonists of Abeta oligomers, reducing the binding of Abeta oligomers to neurons in vitro, preventing spine loss in neurons and preventing and treating oligomer-induced deficits in membrane trafficking. These molecules are highly brain penetrant and prevent and restore cognitive deficits in mouse models of Alzheimer's disease. Counter-screening these compounds against a broad panel of potential CNS targets revealed they are highly potent and specific ligands of the sigma-2/PGRMC1 receptor. Brain concentrations of the compounds corresponding to greater than 80% receptor occupancy at the sigma-2/PGRMC1 receptor restore cognitive function in transgenic hAPP Swe/Ldn mice. These studies demonstrate that synthetic and human-derived Abeta oligomers act as pharmacologically-behaved ligands at neuronal receptors--i.e. they exhibit saturable binding to a target, they exert a functional effect related to their binding and their displacement by small molecule antagonists blocks their functional effect. The first-in-class small molecule receptor antagonists described here restore memory to normal in multiple AD models and sustain improvement long-term, representing a novel mechanism of action for disease-modifying Alzheimer's therapeutics.
Collapse
Affiliation(s)
- Nicholas J. Izzo
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| | - Agnes Staniszewski
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Lillian To
- Stanford University Medical School Behavioral and Functional Neuroscience Laboratory, Palo Alto, California, United States of America
| | - Mauro Fa
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Andrew F. Teich
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Faisal Saeed
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Harrison Wostein
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Thomas Walko
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Anisha Vaswani
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Meghan Wardius
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Zanobia Syed
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Jessica Ravenscroft
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Kelsie Mozzoni
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Colleen Silky
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Courtney Rehak
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Raymond Yurko
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Patricia Finn
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Gary Look
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Gilbert Rishton
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Hank Safferstein
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Miles Miller
- Department of Pathology and Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Conrad Johanson
- Department of Pathology and Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Edward Stopa
- Department of Pathology and Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | | | | | - Mehrdad Shamloo
- Stanford University Medical School Behavioral and Functional Neuroscience Laboratory, Palo Alto, California, United States of America
| | - Ottavio Arancio
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Harry LeVine
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | - Susan M. Catalano
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
45
|
Welzel AT, Maggio JE, Shankar GM, Walker DE, Ostaszewski BL, Li S, Klyubin I, Rowan MJ, Seubert P, Walsh DM, Selkoe DJ. Secreted amyloid β-proteins in a cell culture model include N-terminally extended peptides that impair synaptic plasticity. Biochemistry 2014; 53:3908-21. [PMID: 24840308 PMCID: PMC4070750 DOI: 10.1021/bi5003053] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
![]()
Evidence
for a central role of amyloid β-protein (Aβ) in the genesis
of Alzheimer’s disease (AD) has led to
advanced human trials of Aβ-lowering agents. The “amyloid
hypothesis” of AD postulates deleterious effects of small,
soluble forms of Aβ on synaptic form and function. Because selectively
targeting synaptotoxic forms of soluble Aβ could be therapeutically
advantageous, it is important to understand the full range of soluble
Aβ derivatives. We previously described a Chinese hamster ovary (CHO) cell line (7PA2 cells) that stably expresses mutant human amyloid precursor protein (APP). Here, we extend this work by purifying an sodium dodecyl sulfate
(SDS)-stable, ∼8 kDa Aβ species
from the 7PA2 medium. Mass spectrometry confirmed its identity as
a noncovalently bonded Aβ40 homodimer that impaired hippocampal
long-term potentiation (LTP) in vivo. We further report the detection
of Aβ-containing fragments of APP in the 7PA2 medium that extend
N-terminal from Asp1 of Aβ. These N-terminally extended Aβ-containing
monomeric fragments are distinct from soluble Aβ oligomers formed
from Aβ1-40/42 monomers and are bioactive synaptotoxins secreted
by 7PA2 cells. Importantly, decreasing β-secretase processing
of APP elevated these alternative synaptotoxic APP fragments. We conclude
that certain synaptotoxic Aβ-containing species can arise from
APP processing events N-terminal to the classical β-secretase
cleavage site.
Collapse
|
46
|
Morris GP, Clark IA, Vissel B. Inconsistencies and controversies surrounding the amyloid hypothesis of Alzheimer's disease. Acta Neuropathol Commun 2014; 2:135. [PMID: 25231068 PMCID: PMC4207354 DOI: 10.1186/s40478-014-0135-5] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 08/30/2014] [Indexed: 12/16/2022] Open
Abstract
The amyloid hypothesis has driven drug development strategies for Alzheimer's disease for over 20 years. We review why accumulation of amyloid-beta (Aβ) oligomers is generally considered causal for synaptic loss and neurodegeneration in AD. We elaborate on and update arguments for and against the amyloid hypothesis with new data and interpretations, and consider why the amyloid hypothesis may be failing therapeutically. We note several unresolved issues in the field including the presence of Aβ deposition in cognitively normal individuals, the weak correlation between plaque load and cognition, questions regarding the biochemical nature, presence and role of Aβ oligomeric assemblies in vivo, the bias of pre-clinical AD models toward the amyloid hypothesis and the poorly explained pathological heterogeneity and comorbidities associated with AD. We also illustrate how extensive data cited in support of the amyloid hypothesis, including genetic links to disease, can be interpreted independently of a role for Aβ in AD. We conclude it is essential to expand our view of pathogenesis beyond Aβ and tau pathology and suggest several future directions for AD research, which we argue will be critical to understanding AD pathogenesis.
Collapse
Affiliation(s)
- Gary P Morris
- />Garvan Institute of Medical Research, Neuroscience Department, Neurodegenerative Disorders Laboratory, 384 Victoria Street, Darlinghurst, NSW 2010 Australia
- />Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Ian A Clark
- />Research School of Biology, Australian National University, Canberra, Australia
| | - Bryce Vissel
- />Garvan Institute of Medical Research, Neuroscience Department, Neurodegenerative Disorders Laboratory, 384 Victoria Street, Darlinghurst, NSW 2010 Australia
- />Faculty of Medicine, University of New South Wales, Sydney, Australia
| |
Collapse
|
47
|
Seroprevalence of N-methyl-D-aspartate glutamate receptor (NMDA-R) autoantibodies in aging subjects without neuropsychiatric disorders and in dementia patients. Eur Arch Psychiatry Clin Neurosci 2014; 264:545-50. [PMID: 24604707 DOI: 10.1007/s00406-014-0493-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Accepted: 02/20/2014] [Indexed: 10/25/2022]
Abstract
N-methyl-D-aspartate glutamate receptors (NMDA-R) play a key role in learning and memory. Therefore, they may be involved in the pathophysiology of dementia. NMDA-R autoantibodies directed against the NR1a subunit of the NMDA-R, which were first identified as a specific marker for a severe form of encephalitis, cause a decrease in NMDA-Rs, resulting in cognitive impairment and psychosis. We examined the prevalence of NR1a NMDA-R autoantibodies in the serum and cerebrospinal fluid (CSF) of 24 patients with Alzheimer's disease (AD), 20 patients with subcortical ischemic vascular dementia (SIVD), and 274 volunteers without neuropsychiatric disorder. The latter cases showed an association of seropositivity with age. Notably, the overall seroprevalence was not statistically different between dementia patients and matched controls. Further analysis of the patient samples showed that four patients with AD and three patients with SIVD had positive NMDA-R IgM, IgG, and/or IgA autoantibody titers in serum. These patients suffered from psychosis (with the exception of one case). CSF samples were negative for NMDA-R autoantibodies. We conclude that the seroprevalence of NMDA-R-directed autoantibodies is age-related. It has to be clarified by larger studies whether NMDA-R autoantibodies in peripheral blood may predispose patients with AD and SIVD to susceptibility for psychotic episodes if disturbances of blood-brain-barrier integrity occur.
Collapse
|
48
|
Genetic inhibition of phosphorylation of the translation initiation factor eIF2α does not block Aβ-dependent elevation of BACE1 and APP levels or reduce amyloid pathology in a mouse model of Alzheimer's disease. PLoS One 2014; 9:e101643. [PMID: 24992504 PMCID: PMC4081565 DOI: 10.1371/journal.pone.0101643] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 06/07/2014] [Indexed: 11/30/2022] Open
Abstract
β-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1) initiates the production of β-amyloid (Aβ), the major constituent of amyloid plaques in Alzheimer’s disease (AD). BACE1 is elevated ∼2–3 fold in AD brain and is concentrated in dystrophic neurites near plaques, suggesting BACE1 elevation is Aβ−dependent. Previously, we showed that phosphorylation of the translation initiation factor eIF2α de-represses translation of BACE1 mRNA following stress such as energy deprivation. We hypothesized that stress induced by Aβ might increase BACE1 levels by the same translational mechanism involving eIF2α phosphorylation. To test this hypothesis, we used three different genetic strategies to determine the effects of reducing eIF2α phosphorylation on Aβ-dependent BACE1 elevation in vitro and in vivo: 1) a two-vector adeno-associated virus (AAV) system to express constitutively active GADD34, the regulatory subunit of PP1c eIF2α phosphatase; 2) a non-phosphorylatable eIF2α S51A knockin mutation; 3) a BACE1-YFP transgene lacking the BACE1 mRNA 5′ untranslated region (UTR) required for eIF2α translational regulation. The first two strategies were used in primary neurons and 5XFAD transgenic mice, while the third strategy was employed only in 5XFAD mice. Despite very effective reduction of eIF2α phosphorylation in both primary neurons and 5XFAD brains, or elimination of eIF2α-mediated regulation of BACE1-YFP mRNA translation in 5XFAD brains, Aβ-dependent BACE1 elevation was not decreased. Additionally, robust inhibition of eIF2α phosphorylation did not block Aβ-dependent APP elevation in primary neurons, nor did it reduce amyloid pathology in 5XFAD mice. We conclude that amyloid-associated BACE1 elevation is not caused by translational de-repression via eIF2α phosphorylation, but instead appears to involve a post-translational mechanism. These definitive genetic results exclude a role for eIF2α phosphorylation in Aβ-dependent BACE1 and APP elevation. We suggest a vicious pathogenic cycle wherein Aβ42 toxicity induces peri-plaque BACE1 and APP accumulation in dystrophic neurites leading to exacerbated Aβ production and plaque progression.
Collapse
|
49
|
Busse S, Brix B, Kunschmann R, Bogerts B, Stoecker W, Busse M. N-methyl-d-aspartate glutamate receptor (NMDA-R) antibodies in mild cognitive impairment and dementias. Neurosci Res 2014; 85:58-64. [PMID: 24973618 DOI: 10.1016/j.neures.2014.06.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 06/06/2014] [Accepted: 06/12/2014] [Indexed: 01/07/2023]
Abstract
The N-methyl-d-aspartate glutamate receptor (NMDA-R) plays a central role in learning and memory and has therefore a potential role in the pathophysiology of neuropsychiatric disorders. Recently, we detected NMDA-R autoantibodies in aged healthy volunteers without neuropsychiatric disorders. Since studies showing the involvement of NMDA-R antibodies in mild cognitive impairment and different forms of dementia are rare, we examined NMDA-R antibodies (Abs) in serum of 46 patients with Alzheimer's disease (AD), 26 patients with subcortical ischemic vascular dementia (SIVD), 18 patients with frontotemporal dementia (FTD), 11 patients with Lewy body disease (LBD) and 33 patients with mild cognitive impairment (MCI) and in 21 healthy aged, gender-matched volunteers. While IgM and/or IgA NMDA-R Abs were present in all groups, IgG was only detected in one AD sample. Seropositivity could be correlated with the presence of co-symptoms: MCI and AD patients suffering from depression and AD and SIVD patients with a psychosis were almost all NMDA-R Ab positive. We conclude that the presence of NMDA-R Abs in dementia could influence the incidence of comorbid depressive and/or psychotic states.
Collapse
Affiliation(s)
- Stefan Busse
- Department of Psychiatry, University of Magdeburg, Magdeburg, Germany.
| | - Britta Brix
- Institute for Experimental Immunology, Affiliated to Euroimmun, Lübeck, Lübeck, Germany
| | - Ralf Kunschmann
- Department of Psychiatry, University of Magdeburg, Magdeburg, Germany
| | - Bernhard Bogerts
- Department of Psychiatry, University of Magdeburg, Magdeburg, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Winfried Stoecker
- Institute for Experimental Immunology, Affiliated to Euroimmun, Lübeck, Lübeck, Germany
| | - Mandy Busse
- Department of Pediatric Pulmonology & Allergology, Medical University of Hannover, Hannover, Germany
| |
Collapse
|
50
|
McIntire LBJ, Landman N, Kang MS, Finan GM, Hwang JC, Moore AZ, Park LS, Lin CS, Kim TW. Phenotypic assays for β-amyloid in mouse embryonic stem cell-derived neurons. ACTA ACUST UNITED AC 2014; 20:956-67. [PMID: 23890013 DOI: 10.1016/j.chembiol.2013.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 06/07/2013] [Accepted: 06/13/2013] [Indexed: 10/26/2022]
Abstract
Given the complex nature of Alzheimer's disease (AD), a cell-based model that recapitulates the physiological properties of the target neuronal population would be extremely valuable for discovering improved drug candidates and chemical probes to uncover disease mechanisms. We established phenotypic neuronal assays for the biogenesis and synaptic action of amyloid β peptide (Aβ) based on embryonic stem cell-derived neurons (ESNs). ESNs enriched with pyramidal neurons were robust, scalable, and amenable to a small-molecule screening assay, overcoming the apparent limitations of neuronal models derived from human pluripotent cells. Small-molecule screening of clinical compounds identified four compounds capable of reducing Aβ levels in ESNs derived from the Tg2576 mouse model of AD. Our approach is therefore highly suitable for phenotypic screening in AD drug discovery and has the potential to identify therapeutic candidates with improved efficacy and safety potential.
Collapse
Affiliation(s)
- Laura Beth J McIntire
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | |
Collapse
|