1
|
Latif‐Hernandez A, Yang T, Butler RR, Losada PM, Minhas PS, White H, Tran KC, Liu H, Simmons DA, Langness V, Andreasson KI, Wyss‐Coray T, Longo FM. A TrkB and TrkC partial agonist restores deficits in synaptic function and promotes activity-dependent synaptic and microglial transcriptomic changes in a late-stage Alzheimer's mouse model. Alzheimers Dement 2024; 20:4434-4460. [PMID: 38779814 PMCID: PMC11247716 DOI: 10.1002/alz.13857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 03/12/2024] [Accepted: 04/02/2024] [Indexed: 05/25/2024]
Abstract
INTRODUCTION Tropomyosin related kinase B (TrkB) and C (TrkC) receptor signaling promotes synaptic plasticity and interacts with pathways affected by amyloid beta (Aβ) toxicity. Upregulating TrkB/C signaling could reduce Alzheimer's disease (AD)-related degenerative signaling, memory loss, and synaptic dysfunction. METHODS PTX-BD10-2 (BD10-2), a small molecule TrkB/C receptor partial agonist, was orally administered to aged London/Swedish-APP mutant mice (APPL/S) and wild-type controls. Effects on memory and hippocampal long-term potentiation (LTP) were assessed using electrophysiology, behavioral studies, immunoblotting, immunofluorescence staining, and RNA sequencing. RESULTS In APPL/S mice, BD10-2 treatment improved memory and LTP deficits. This was accompanied by normalized phosphorylation of protein kinase B (Akt), calcium-calmodulin-dependent kinase II (CaMKII), and AMPA-type glutamate receptors containing the subunit GluA1; enhanced activity-dependent recruitment of synaptic proteins; and increased excitatory synapse number. BD10-2 also had potentially favorable effects on LTP-dependent complement pathway and synaptic gene transcription. DISCUSSION BD10-2 prevented APPL/S/Aβ-associated memory and LTP deficits, reduced abnormalities in synapse-related signaling and activity-dependent transcription of synaptic genes, and bolstered transcriptional changes associated with microglial immune response. HIGHLIGHTS Small molecule modulation of tropomyosin related kinase B (TrkB) and C (TrkC) restores long-term potentiation (LTP) and behavior in an Alzheimer's disease (AD) model. Modulation of TrkB and TrkC regulates synaptic activity-dependent transcription. TrkB and TrkC receptors are candidate targets for translational therapeutics. Electrophysiology combined with transcriptomics elucidates synaptic restoration. LTP identifies neuron and microglia AD-relevant human-mouse co-expression modules.
Collapse
Affiliation(s)
- Amira Latif‐Hernandez
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Tao Yang
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Robert R. Butler
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Patricia Moran Losada
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordCaliforniaUSA
| | - Paras S. Minhas
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Halle White
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Kevin C. Tran
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Harry Liu
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Danielle A. Simmons
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Vanessa Langness
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Katrin I. Andreasson
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordCaliforniaUSA
- Chan Zuckerberg BiohubSan FranciscoCaliforniaUSA
| | - Tony Wyss‐Coray
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordCaliforniaUSA
- The Phil and Penny Knight Initiative for Brain ResilienceStanford UniversityStanfordCaliforniaUSA
| | - Frank M. Longo
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordCaliforniaUSA
| |
Collapse
|
2
|
Latif-Hernandez A, Yang T, Raymond-Butler R, Losada PM, Minhas P, White H, Tran KC, Liu H, Simmons DA, Langness V, Andreasson K, Wyss-Coray T, Longo FM. A TrkB and TrkC partial agonist restores deficits in synaptic function and promotes activity-dependent synaptic and microglial transcriptomic changes in a late-stage Alzheimer's mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.18.558138. [PMID: 37781573 PMCID: PMC10541128 DOI: 10.1101/2023.09.18.558138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Introduction TrkB and TrkC receptor signaling promotes synaptic plasticity and interacts with pathways affected by amyloid-β (Aβ)-toxicity. Upregulating TrkB/C signaling could reduce Alzheimer's disease (AD)-related degenerative signaling, memory loss, and synaptic dysfunction. Methods PTX-BD10-2 (BD10-2), a small molecule TrkB/C receptor partial agonist, was orally administered to aged London/Swedish-APP mutant mice (APP L/S ) and wild-type controls (WT). Effects on memory and hippocampal long-term potentiation (LTP) were assessed using electrophysiology, behavioral studies, immunoblotting, immunofluorescence staining, and RNA-sequencing. Results Memory and LTP deficits in APP L/S mice were attenuated by treatment with BD10-2. BD10-2 prevented aberrant AKT, CaMKII, and GLUA1 phosphorylation, and enhanced activity-dependent recruitment of synaptic proteins. BD10-2 also had potentially favorable effects on LTP-dependent complement pathway and synaptic gene transcription. Conclusions BD10-2 prevented APP L/S /Aβ-associated memory and LTP deficits, reduced abnormalities in synapse-related signaling and activity-dependent transcription of synaptic genes, and bolstered transcriptional changes associated with microglial immune response.
Collapse
|
3
|
Wood JI, Wong E, Joghee R, Balbaa A, Vitanova KS, Stringer KM, Vanshoiack A, Phelan SLJ, Launchbury F, Desai S, Tripathi T, Hanrieder J, Cummings DM, Hardy J, Edwards FA. Plaque contact and unimpaired Trem2 is required for the microglial response to amyloid pathology. Cell Rep 2022; 41:111686. [PMID: 36417868 DOI: 10.1016/j.celrep.2022.111686] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 08/30/2022] [Accepted: 10/27/2022] [Indexed: 11/23/2022] Open
Abstract
Using spatial cell-type-enriched transcriptomics, we compare plaque-induced gene (PIG) expression in microglia-touching plaques, neighboring plaques, and far from plaques in an aged Alzheimer's mouse model with late plaque development. In 18-month-old APPNL-F/NL-F knockin mice, with and without the Alzheimer's disease risk mutation Trem2R47H/R47H, we report that expression of 38/55 PIGs have plaque-induced microglial upregulation, with a subset only upregulating in microglia directly contacting plaques. For seven PIGs, including Trem2, this upregulation is prevented in APPNL-F/NL-FTrem2R47H/R47H mice. These TREM2-dependent genes are all involved in phagocytic and degradative processes that we show correspond to a decrease in phagocytic markers and an increase in the density of small plaques in Trem2-mutated mice. Furthermore, despite the R47H mutation preventing increased Trem2 gene expression, TREM2 protein levels and microglial density are still marginally increased on plaques. Hence, both microglial contact with plaques and functioning TREM2 are necessary for microglia to respond appropriately to amyloid pathology.
Collapse
Affiliation(s)
- Jack I Wood
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V3, 43180 Mölndal, Sweden
| | - Eugenia Wong
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Ridwaan Joghee
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Aya Balbaa
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Karina S Vitanova
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Katie M Stringer
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V3, 43180 Mölndal, Sweden
| | - Alison Vanshoiack
- Nanostring Technologies, 530 Fairview Avenue N, Seattle, WA 98109, United States
| | | | - Francesca Launchbury
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Sneha Desai
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Takshashila Tripathi
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Jörg Hanrieder
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V3, 43180 Mölndal, Sweden
| | - Damian M Cummings
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - John Hardy
- Dementia Research Institute, University College London, Gower Street, London WC1E 6BT, UK; Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Frances A Edwards
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK; Institute of Healthy Ageing, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
4
|
Neuwirth LS, Verrengia MT, Harikinish-Murrary ZI, Orens JE, Lopez OE. Under or Absent Reporting of Light Stimuli in Testing of Anxiety-Like Behaviors in Rodents: The Need for Standardization. Front Mol Neurosci 2022; 15:912146. [PMID: 36061362 PMCID: PMC9428565 DOI: 10.3389/fnmol.2022.912146] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
Behavioral neuroscience tests such as the Light/Dark Test, the Open Field Test, the Elevated Plus Maze Test, and the Three Chamber Social Interaction Test have become both essential and widely used behavioral tests for transgenic and pre-clinical models for drug screening and testing. However, as fast as the field has evolved and the contemporaneous involvement of technology, little assessment of the literature has been done to ensure that these behavioral neuroscience tests that are crucial to pre-clinical testing have well-controlled ethological motivation by the use of lighting (i.e., Lux). In the present review paper, N = 420 manuscripts were examined from 2015 to 2019 as a sample set (i.e., n = ~20–22 publications per year) and it was found that only a meager n = 50 publications (i.e., 11.9% of the publications sampled) met the criteria for proper anxiogenic and anxiolytic Lux reported. These findings illustrate a serious concern that behavioral neuroscience papers are not being vetted properly at the journal review level and are being released into the literature and public domain making it difficult to assess the quality of the science being reported. This creates a real need for standardizing the use of Lux in all publications on behavioral neuroscience techniques within the field to ensure that contributions are meaningful, avoid unnecessary duplication, and ultimately would serve to create a more efficient process within the pre-clinical screening/testing for drugs that serve as anxiolytic compounds that would prove more useful than what prior decades of work have produced. It is suggested that improving the standardization of the use and reporting of Lux in behavioral neuroscience tests and the standardization of peer-review processes overseeing the proper documentation of these methodological approaches in manuscripts could serve to advance pre-clinical testing for effective anxiolytic drugs. This report serves to highlight this concern and proposes strategies to proactively remedy them as the field moves forward for decades to come.
Collapse
Affiliation(s)
- Lorenz S. Neuwirth
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
- *Correspondence: Lorenz S. Neuwirth
| | - Michael T. Verrengia
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
| | - Zachary I. Harikinish-Murrary
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
| | - Jessica E. Orens
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
| | - Oscar E. Lopez
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
| |
Collapse
|
5
|
Kumar SP, Babu PP. NADPH Oxidase: a Possible Therapeutic Target for Cognitive Impairment in Experimental Cerebral Malaria. Mol Neurobiol 2021; 59:800-820. [PMID: 34782951 DOI: 10.1007/s12035-021-02598-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/12/2021] [Indexed: 12/19/2022]
Abstract
Long-term cognitive impairment associated with seizure-induced hippocampal damage is the key feature of cerebral malaria (CM) pathogenesis. One-fourth of child survivors of CM suffer from long-lasting neurological deficits and behavioral anomalies. However, mechanisms on hippocampal dysfunction are unclear. In this study, we elucidated whether gp91phox isoform of nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2) (a potent marker of oxidative stress) mediates hippocampal neuronal abnormalities and cognitive dysfunction in experimental CM (ECM). Mice symptomatic to CM were rescue treated with artemether monotherapy (ARM) and in combination with apocynin (ARM + APO) adjunctive based on scores of Rapid Murine Come behavior Scale (RMCBS). After a 30-day survivability period, we performed Barnes maze, T-maze, and novel object recognition cognitive tests to evaluate working and reference memory in all the experimental groups except CM. Sensorimotor tests were conducted in all the cohorts to assess motor coordination. We performed Golgi-Cox staining to illustrate cornu ammonis-1 (CA1) pyramidal neuronal morphology and study overall hippocampal neuronal density changes. Further, expression of NOX2, NeuN (neuronal marker) in hippocampal CA1 and dentate gyrus was determined using double immunofluorescence experiments in all the experimental groups. Mice administered with ARM monotherapy and APO adjunctive treatment exhibited similar survivability. The latter showed better locomotor and cognitive functions, reduced ROS levels, and hippocampal NOX2 immunoreactivity in ECM. Our results show a substantial increase in hippocampal NeuN immunoreactivity and dendritic arborization in ARM + APO cohorts compared to ARM-treated brain samples. Overall, our study suggests that overexpression of NOX2 could result in loss of hippocampal neuronal density and dendritic spines of CA1 neurons affecting the spatial working and reference memory during ECM. Notably, ARM + APO adjunctive therapy reversed the altered neuronal morphology and oxidative damage in hippocampal neurons restoring long-term cognitive functions after CM.
Collapse
Affiliation(s)
- Simhadri Praveen Kumar
- F-23/71, Neuroscience Laboratory, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500 046, India
| | - Phanithi Prakash Babu
- F-23/71, Neuroscience Laboratory, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500 046, India.
| |
Collapse
|
6
|
Benitez DP, Jiang S, Wood J, Wang R, Hall CM, Peerboom C, Wong N, Stringer KM, Vitanova KS, Smith VC, Joshi D, Saito T, Saido TC, Hardy J, Hanrieder J, De Strooper B, Salih DA, Tripathi T, Edwards FA, Cummings DM. Knock-in models related to Alzheimer's disease: synaptic transmission, plaques and the role of microglia. Mol Neurodegener 2021; 16:47. [PMID: 34266459 PMCID: PMC8281661 DOI: 10.1186/s13024-021-00457-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 05/26/2021] [Indexed: 11/18/2022] Open
Abstract
Background Microglia are active modulators of Alzheimer’s disease but their role in relation to amyloid plaques and synaptic changes due to rising amyloid beta is unclear. We add novel findings concerning these relationships and investigate which of our previously reported results from transgenic mice can be validated in knock-in mice, in which overexpression and other artefacts of transgenic technology are avoided. Methods AppNL-F and AppNL-G-F knock-in mice expressing humanised amyloid beta with mutations in App that cause familial Alzheimer’s disease were compared to wild type mice throughout life. In vitro approaches were used to understand microglial alterations at the genetic and protein levels and synaptic function and plasticity in CA1 hippocampal neurones, each in relationship to both age and stage of amyloid beta pathology. The contribution of microglia to neuronal function was further investigated by ablating microglia with CSF1R inhibitor PLX5622. Results Both App knock-in lines showed increased glutamate release probability prior to detection of plaques. Consistent with results in transgenic mice, this persisted throughout life in AppNL-F mice but was not evident in AppNL-G-F with sparse plaques. Unlike transgenic mice, loss of spontaneous excitatory activity only occurred at the latest stages, while no change could be detected in spontaneous inhibitory synaptic transmission or magnitude of long-term potentiation. Also, in contrast to transgenic mice, the microglial response in both App knock-in lines was delayed until a moderate plaque load developed. Surviving PLX5266-depleted microglia tended to be CD68-positive. Partial microglial ablation led to aged but not young wild type animals mimicking the increased glutamate release probability in App knock-ins and exacerbated the App knock-in phenotype. Complete ablation was less effective in altering synaptic function, while neither treatment altered plaque load. Conclusions Increased glutamate release probability is similar across knock-in and transgenic mouse models of Alzheimer’s disease, likely reflecting acute physiological effects of soluble amyloid beta. Microglia respond later to increased amyloid beta levels by proliferating and upregulating Cd68 and Trem2. Partial depletion of microglia suggests that, in wild type mice, alteration of surviving phagocytic microglia, rather than microglial loss, drives age-dependent effects on glutamate release that become exacerbated in Alzheimer’s disease. Supplementary Information The online version contains supplementary material available at 10.1186/s13024-021-00457-0.
Collapse
Affiliation(s)
- Diana P Benitez
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Shenyi Jiang
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.,Ludwig Maximilians Universitat, Munich, Germany
| | - Jack Wood
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Rui Wang
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.,Institute for Synaptic Physiology, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Chloe M Hall
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.,School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK
| | - Carlijn Peerboom
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.,Cell Biology, Neurobiology and Biophysics, Biology Department, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Natalie Wong
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Katie M Stringer
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Karina S Vitanova
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Victoria C Smith
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.,Centre for Doctoral Training at the Institute of Health Informatics, University College London, 222 Euston Road, London, NW1 2DA, UK
| | - Dhaval Joshi
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.,Department of Psychology, University of Cambridge, Cambridge, UK
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Wako-shi, Saitama, 351-0198, Japan.,Department of Neurocognitive Science, Institute of Brain Science, Nagoya City, University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Wako-shi, Saitama, 351-0198, Japan
| | - John Hardy
- Dementia Research Institute, University College London, Gower Street, London, WC1E 6BT, UK.,Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK.,UCL Movement Disorders Centre, University College London, London, UK.,Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong, SAR, China
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Bart De Strooper
- Dementia Research Institute, University College London, Gower Street, London, WC1E 6BT, UK.,VIB Center for Brain & Disease Research, 3000, Leuven, KU, Belgium.,Department of Neurosciences, Leuven Brain Institute, 3000, Leuven, Belgium
| | - Dervis A Salih
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.,Dementia Research Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Takshashila Tripathi
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Frances A Edwards
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK. .,Institute of Healthy Ageing, University College London, Gower Street, London, WC1E 6BT, UK.
| | - Damian M Cummings
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK. .,Dementia Research Institute, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
7
|
Chaney AM, Lopez-Picon FR, Serrière S, Wang R, Bochicchio D, Webb SD, Vandesquille M, Harte MK, Georgiadou C, Lawrence C, Busson J, Vercouillie J, Tauber C, Buron F, Routier S, Reekie T, Snellman A, Kassiou M, Rokka J, Davies KE, Rinne JO, Salih DA, Edwards FA, Orton LD, Williams SR, Chalon S, Boutin H. Prodromal neuroinflammatory, cholinergic and metabolite dysfunction detected by PET and MRS in the TgF344-AD transgenic rat model of AD: a collaborative multi-modal study. Am J Cancer Res 2021; 11:6644-6667. [PMID: 34093845 PMCID: PMC8171096 DOI: 10.7150/thno.56059] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/15/2021] [Indexed: 12/25/2022] Open
Abstract
Mouse models of Alzheimer's disease (AD) are valuable but do not fully recapitulate human AD pathology, such as spontaneous Tau fibril accumulation and neuronal loss, necessitating the development of new AD models. The transgenic (TG) TgF344-AD rat has been reported to develop age-dependent AD features including neuronal loss and neurofibrillary tangles, despite only expressing APP and PSEN1 mutations, suggesting an improved modelling of AD hallmarks. Alterations in neuronal networks as well as learning performance and cognition tasks have been reported in this model, but none have combined a longitudinal, multimodal approach across multiple centres, which mimics the approaches commonly taken in clinical studies. We therefore aimed to further characterise the progression of AD-like pathology and cognition in the TgF344-AD rat from young-adults (6 months (m)) to mid- (12 m) and advanced-stage (18 m, 25 m) of the disease. Methods: TgF344-AD rats and wild-type (WT) littermates were imaged at 6 m, 12 m and 18 m with [18F]DPA-714 (TSPO, neuroinflammation), [18F]Florbetaben (Aβ) and [18F]ASEM (α7-nicotinic acetylcholine receptor) and with magnetic resonance spectroscopy (MRS) and with (S)-[18F]THK5117 (Tau) at 15 and 25 m. Behaviour tests were also performed at 6 m, 12 m and 18 m. Immunohistochemistry (CD11b, GFAP, Aβ, NeuN, NeuroChrom) and Tau (S)-[18F]THK5117 autoradiography, immunohistochemistry and Western blot were also performed. Results: [18F]DPA-714 positron emission tomography (PET) showed an increase in neuroinflammation in TG vs wildtype animals from 12 m in the hippocampus (+11%), and at the advanced-stage AD in the hippocampus (+12%), the thalamus (+11%) and frontal cortex (+14%). This finding coincided with strong increases in brain microgliosis (CD11b) and astrogliosis (GFAP) at these time-points as assessed by immunohistochemistry. In vivo [18F]ASEM PET revealed an age-dependent increase uptake in the striatum and pallidum/nucleus basalis of Meynert in WT only, similar to that observed with this tracer in humans, resulting in TG being significantly lower than WT by 18 m. In vivo [18F]Florbetaben PET scanning detected Aβ accumulation at 18 m, and (S)-[18F]THK5117 PET revealed subsequent Tau accumulation at 25m in hippocampal and cortical regions. Aβ plaques were low but detectable by immunohistochemistry from 6 m, increasing further at 12 and 18 m with Tau-positive neurons adjacent to Aβ plaques at 18 m. NeuroChrom (a pan neuronal marker) immunohistochemistry revealed a loss of neuronal staining at the Aβ plaques locations, while NeuN labelling revealed an age-dependent decrease in hippocampal neuron number in both genotypes. Behavioural assessment using the novel object recognition task revealed that both WT & TgF344-AD animals discriminated the novel from familiar object at 3 m and 6 m of age. However, low levels of exploration observed in both genotypes at later time-points resulted in neither genotype successfully completing the task. Deficits in social interaction were only observed at 3 m in the TgF344-AD animals. By in vivo MRS, we showed a decrease in neuronal marker N-acetyl-aspartate in the hippocampus at 18 m (-18% vs age-matched WT, and -31% vs 6 m TG) and increased Taurine in the cortex of TG (+35% vs age-matched WT, and +55% vs 6 m TG). Conclusions: This multi-centre multi-modal study demonstrates, for the first time, alterations in brain metabolites, cholinergic receptors and neuroinflammation in vivo in this model, validated by robust ex vivo approaches. Our data confirm that, unlike mouse models, the TgF344-AD express Tau pathology that can be detected via PET, albeit later than by ex vivo techniques, and is a useful model to assess and longitudinally monitor early neurotransmission dysfunction and neuroinflammation in AD.
Collapse
|
8
|
Tzimourta KD, Christou V, Tzallas AT, Giannakeas N, Astrakas LG, Angelidis P, Tsalikakis D, Tsipouras MG. Machine Learning Algorithms and Statistical Approaches for Alzheimer's Disease Analysis Based on Resting-State EEG Recordings: A Systematic Review. Int J Neural Syst 2021; 31:2130002. [PMID: 33588710 DOI: 10.1142/s0129065721300023] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder and the most common type of dementia with a great prevalence in western countries. The diagnosis of AD and its progression is performed through a variety of clinical procedures including neuropsychological and physical examination, Electroencephalographic (EEG) recording, brain imaging and blood analysis. During the last decades, analysis of the electrophysiological dynamics in AD patients has gained great research interest, as an alternative and cost-effective approach. This paper summarizes recent publications focusing on (a) AD detection and (b) the correlation of quantitative EEG features with AD progression, as it is estimated by Mini Mental State Examination (MMSE) score. A total of 49 experimental studies published from 2009 until 2020, which apply machine learning algorithms on resting state EEG recordings from AD patients, are reviewed. Results of each experimental study are presented and compared. The majority of the studies focus on AD detection incorporating Support Vector Machines, while deep learning techniques have not yet been applied on large EEG datasets. Promising conclusions for future studies are presented.
Collapse
Affiliation(s)
- Katerina D Tzimourta
- Department of Electrical and Computer Engineering, University of Western Macedonia, Kozani, GR50100, Greece
- Department of Medical Physics, Medical School, University of Ioannina, Ioannina GR45110, Greece
| | - Vasileios Christou
- Q Base R&D, Science & Technology Park of Epirus, University of Ioannina Campus, Ioannina GR45110, Greece
- Department of Informatics and Telecommunications, School of Informatics and Telecommunications, University of Ioannina, Arta GR47100, Greece
| | - Alexandros T Tzallas
- Department of Informatics and Telecommunications, School of Informatics and Telecommunications, University of Ioannina, Arta GR47100, Greece
| | - Nikolaos Giannakeas
- Department of Informatics and Telecommunications, School of Informatics and Telecommunications, University of Ioannina, Arta GR47100, Greece
| | - Loukas G Astrakas
- Department of Medical Physics, Medical School, University of Ioannina, Ioannina GR45110, Greece
| | - Pantelis Angelidis
- Department of Electrical and Computer Engineering, University of Western Macedonia, Kozani GR50100, Greece
| | - Dimitrios Tsalikakis
- Department of Electrical and Computer Engineering, University of Western Macedonia, Kozani GR50100, Greece
| | - Markos G Tsipouras
- Department of Electrical and Computer Engineering, University of Western Macedonia, Kozani GR50100, Greece
| |
Collapse
|
9
|
A Unifying Hypothesis for Alzheimer's Disease: From Plaques to Neurodegeneration. Trends Neurosci 2020; 42:310-322. [PMID: 31006494 DOI: 10.1016/j.tins.2019.03.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 12/21/2022]
Abstract
Evidence suggests that amyloid β is highly toxic to synapses in a phospho-Tau-dependent manner. Here, I present a hypothesis that links previous evidence from the first rise of amyloid β through to Tau tangles and neurodegeneration. In the immediate vicinity of plaques, concentrated soluble amyloid β occurs in equilibrium with deposited forms. Initially, plaques cover only a small percentage of brain volume. Microglia, by efficiently removing damaged synapses, may prevent spread of damage along the axon, restricting damage to the immediate vicinity of plaques. However, as plaque load increases, as seen in Alzheimer's disease, an individual axon may suffer multiple points of damage, leading to dissociation of Tau, formation of a tangle, and loss of the axon. As more axons suffer this fate, the network eventually degenerates. According to this hypothesis, the degree of plaque load that an individual can tolerate would depend on the efficiency of their microglia in removing amyloid-β-damaged synapses and the distribution of plaques, relative to axon trajectories, would determine the eventual cognitive symptoms.
Collapse
|
10
|
Sos KE, Mayer MI, Takács VT, Major A, Bardóczi Z, Beres BM, Szeles T, Saito T, Saido TC, Mody I, Freund TF, Nyiri G. Amyloid β induces interneuron-specific changes in the hippocampus of APPNL-F mice. PLoS One 2020; 15:e0233700. [PMID: 32469963 PMCID: PMC7259556 DOI: 10.1371/journal.pone.0233700] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 05/11/2020] [Indexed: 01/07/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline and amyloid-beta (Aβ) depositions generated by the proteolysis of amyloid precursor protein (APP) in the brain. In APPNL-F mice, APP gene was humanized and contains two familial AD mutations, and APP-unlike other mouse models of AD-is driven by the endogenous mouse APP promoter. Similar to people without apparent cognitive dysfunction but with heavy Aβ plaque load, we found no significant decline in the working memory of adult APPNL-F mice, but these mice showed decline in the expression of normal anxiety. Using immunohistochemistry and 3D block-face scanning electron microscopy, we found no changes in GABAA receptor positivity and size of somatic and dendritic synapses of hippocampal interneurons. We did not find alterations in the level of expression of perineuronal nets around parvalbumin (PV) interneurons or in the density of PV- or somatostatin-positive hippocampal interneurons. However, in contrast to other investigated cell types, PV interneuron axons were occasionally mildly dystrophic around Aβ plaques, and the synapses of PV-positive axon initial segment (AIS)-targeting interneurons were significantly enlarged. Our results suggest that PV interneurons are highly resistant to amyloidosis in APPNL-F mice and amyloid-induced increase in hippocampal pyramidal cell excitability may be compensated by PV-positive AIS-targeting cells. Mechanisms that make PV neurons more resilient could therefore be exploited in the treatment of AD for mitigating Aβ-related inflammatory effects on neurons.
Collapse
Affiliation(s)
- Katalin E. Sos
- Department of Cellular and Network Neurobiology, Institute of Experimental Medicine, HAS, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Márton I. Mayer
- Department of Cellular and Network Neurobiology, Institute of Experimental Medicine, HAS, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Virág T. Takács
- Department of Cellular and Network Neurobiology, Institute of Experimental Medicine, HAS, Budapest, Hungary
| | - Abel Major
- Department of Cellular and Network Neurobiology, Institute of Experimental Medicine, HAS, Budapest, Hungary
| | - Zsuzsanna Bardóczi
- Department of Cellular and Network Neurobiology, Institute of Experimental Medicine, HAS, Budapest, Hungary
| | - Barnabas M. Beres
- Department of Cellular and Network Neurobiology, Institute of Experimental Medicine, HAS, Budapest, Hungary
| | - Tamás Szeles
- Department of Cellular and Network Neurobiology, Institute of Experimental Medicine, HAS, Budapest, Hungary
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN, Center for Brain Science, Saitama, Japan
- Department of Neurocognitive Science, Nagoya City University Graduate School of Medical Science, Aichi, Japan
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, RIKEN, Center for Brain Science, Saitama, Japan
| | - István Mody
- Department of Neurology, University of California, Los Angeles, California, United States of America
| | - Tamás F. Freund
- Department of Cellular and Network Neurobiology, Institute of Experimental Medicine, HAS, Budapest, Hungary
| | - Gábor Nyiri
- Department of Cellular and Network Neurobiology, Institute of Experimental Medicine, HAS, Budapest, Hungary
| |
Collapse
|
11
|
Salih DA, Bayram S, Guelfi S, Reynolds RH, Shoai M, Ryten M, Brenton JW, Zhang D, Matarin M, Botia JA, Shah R, Brookes KJ, Guetta-Baranes T, Morgan K, Bellou E, Cummings DM, Escott-Price V, Hardy J. Genetic variability in response to amyloid beta deposition influences Alzheimer's disease risk. Brain Commun 2019; 1:fcz022. [PMID: 32274467 PMCID: PMC7145452 DOI: 10.1093/braincomms/fcz022] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 12/18/2022] Open
Abstract
Genome-wide association studies of late-onset Alzheimer's disease risk have previously identified genes primarily expressed in microglia that form a transcriptional network. Using transgenic mouse models of amyloid deposition, we previously showed that many of the mouse orthologues of these risk genes are co-expressed and associated with amyloid pathology. In this new study, we generate an improved RNA-seq-derived network that is expressed in amyloid-responsive mouse microglia and we statistically compare this with gene-level variation in previous human Alzheimer's disease genome-wide association studies to predict at least four new risk genes for the disease (OAS1, LAPTM5, ITGAM/CD11b and LILRB4). Of the mouse orthologues of these genes Oas1a is likely to respond directly to amyloid at the transcriptional level, similarly to established risk gene Trem2, because the increase in Oas1a and Trem2 transcripts in response to amyloid deposition in transgenic mice is significantly higher than both the increase of the average microglial transcript and the increase in microglial number. In contrast, the mouse orthologues of LAPTM5, ITGAM/CD11b and LILRB4 (Laptm5, Itgam/CD11b and Lilra5) show increased transcripts in the presence of amyloid plaques similar in magnitude to the increase of the average microglial transcript and the increase in microglia number, except that Laptm5 and Lilra5 transcripts increase significantly quicker than the average microglial transcript as the plaque load becomes dense. This work suggests that genetic variability in the microglial response to amyloid deposition is a major determinant for Alzheimer's disease risk, and identification of these genes may help to predict the risk of developing Alzheimer's disease. These findings also provide further insights into the mechanisms underlying Alzheimer's disease for potential drug discovery.
Collapse
Affiliation(s)
- Dervis A Salih
- Department of Neuroscience, Physiology and Pharmacology, UCL, Gower Street, London WC1E 6BT, UK
- UK Dementia Research Institute at UCL, Gower Street, London WC1E 6BT, UK
| | | | - Sebastian Guelfi
- Department of Neurodegenerative Diseases, Institute of Neurology, UCL, 1 Wakefield Street, London WC1N 1PJ, UK
| | - Regina H Reynolds
- Department of Neurodegenerative Diseases, Institute of Neurology, UCL, 1 Wakefield Street, London WC1N 1PJ, UK
| | - Maryam Shoai
- UK Dementia Research Institute at UCL, Gower Street, London WC1E 6BT, UK
- Department of Neurodegenerative Diseases, Institute of Neurology, UCL, 1 Wakefield Street, London WC1N 1PJ, UK
| | - Mina Ryten
- Department of Neurodegenerative Diseases, Institute of Neurology, UCL, 1 Wakefield Street, London WC1N 1PJ, UK
| | - Jonathan W Brenton
- Department of Neuroscience, Physiology and Pharmacology, UCL, Gower Street, London WC1E 6BT, UK
| | - David Zhang
- Department of Neurodegenerative Diseases, Institute of Neurology, UCL, 1 Wakefield Street, London WC1N 1PJ, UK
| | - Mar Matarin
- Department of Neurodegenerative Diseases, Institute of Neurology, UCL, 1 Wakefield Street, London WC1N 1PJ, UK
| | - Juan A Botia
- Department of Neurodegenerative Diseases, Institute of Neurology, UCL, 1 Wakefield Street, London WC1N 1PJ, UK
- Department of Information and Communications Engineering, Universidad de Murcia, Spain
| | - Runil Shah
- Department of Neurodegenerative Diseases, Institute of Neurology, UCL, 1 Wakefield Street, London WC1N 1PJ, UK
| | - Keeley J Brookes
- Human Genetics, School of Life Sciences, Life Sciences Building, University Park, University of Nottingham, Nottingham NG7 2RD, UK
| | - Tamar Guetta-Baranes
- Human Genetics, School of Life Sciences, Life Sciences Building, University Park, University of Nottingham, Nottingham NG7 2RD, UK
| | - Kevin Morgan
- Human Genetics, School of Life Sciences, Life Sciences Building, University Park, University of Nottingham, Nottingham NG7 2RD, UK
| | - Eftychia Bellou
- Dementia Research Institute, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK
| | - Damian M Cummings
- Department of Neuroscience, Physiology and Pharmacology, UCL, Gower Street, London WC1E 6BT, UK
| | - Valentina Escott-Price
- Dementia Research Institute, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK
| | - John Hardy
- UK Dementia Research Institute at UCL, Gower Street, London WC1E 6BT, UK
- Department of Neurodegenerative Diseases, Institute of Neurology, UCL, 1 Wakefield Street, London WC1N 1PJ, UK
| |
Collapse
|
12
|
Vitanova KS, Stringer KM, Benitez DP, Brenton J, Cummings DM. Dementia associated with disorders of the basal ganglia. J Neurosci Res 2019; 97:1728-1741. [PMID: 31392765 DOI: 10.1002/jnr.24508] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/19/2019] [Accepted: 07/22/2019] [Indexed: 01/12/2023]
Abstract
Dementia is now the leading cause of death in the United Kingdom, accounting for over 12% of all deaths and is the fifth most common cause of death worldwide. As treatments for heart disease and cancers improve and the population ages, the number of sufferers will only increase, with the chance of developing dementia doubling every 5 years after the age of 65. Finding an effective treatment is ever more critical to avert this pandemic health (and economic) crisis. To date, most dementia-related research has focused on the cortex and the hippocampus; however, with dementia becoming more fully recognized as aspects of diseases historically categorized as motor disorders (e.g., Parkinson's and Huntington's diseases), the role of the basal ganglia in dementia is coming to the fore. Conversely, it is highly likely that neuronal pathways in these structures traditionally considered as spared in Alzheimer's disease are also affected, particularly in later stages of the disease. In this review, we examine some of the limited evidence linking the basal ganglia to dementia.
Collapse
Affiliation(s)
- Karina S Vitanova
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| | - Katie M Stringer
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK.,Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Diana P Benitez
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| | - Jonathan Brenton
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| | - Damian M Cummings
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| |
Collapse
|
13
|
Novel neuroprotective tetramethylpyrazine analog T-006 promotes neurogenesis and neurological restoration in a rat model of stroke. Neuroreport 2019; 30:658-663. [DOI: 10.1097/wnr.0000000000001256] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
14
|
Joel Z, Izquierdo P, Salih DA, Richardson JC, Cummings DM, Edwards FA. Improving Mouse Models for Dementia. Are All the Effects in Tau Mouse Models Due to Overexpression? COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2019; 83:151-161. [PMID: 30745408 DOI: 10.1101/sqb.2018.83.037531] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mouse models of Alzheimer's disease have commonly used transgenic overexpression of genes involved in production of amyloid β (APP and/or PSEN1/2) or Tau (MAPT) with mutations that result in familial forms of dementia. We discuss possible improvements that may create full models while avoiding the problems of overexpression and report synaptic results in APPKI models. We stress use of inappropriate controls without overexpression of the normal human protein and the mismatch between the learning deficits reported in mice with plaques but no tangles and the human condition. We focus on Tau overexpression, including new data that support previous reports of the grossly nonlinear relationship between Tau overexpression and neurofibrillary tangle load, with a twofold increase in Tau protein, resulting in a 100-fold increase in tangle density. These data also support the hypothesis that a high concentration of soluble Tau, in overexpression models, plays an important direct role in neurodegeneration, rather than only via aggregation. Finally, we hypothesize that there is an optimal concentration range over which Tau can bind to microtubules and a threshold beyond which much of the overexpressed protein is unable to bind. The excess thus causes toxicity in ways not necessarily related to the process in human dementias.
Collapse
Affiliation(s)
- Zelah Joel
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, United Kingdom
| | - Pablo Izquierdo
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, United Kingdom
| | - Dervis A Salih
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, United Kingdom
| | - Jill C Richardson
- Neurosciences Therapeutic Area, GlaxoSmithKline R&D, Stevenage, SG1 2NY, United Kingdom
| | - Damian M Cummings
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, United Kingdom
| | - Frances A Edwards
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, United Kingdom
| |
Collapse
|
15
|
Hölscher C. Moving towards a more realistic concept of what constitutes Alzheimer's disease. EBioMedicine 2018; 39:17-18. [PMID: 30579866 PMCID: PMC6355438 DOI: 10.1016/j.ebiom.2018.12.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 12/14/2018] [Indexed: 11/20/2022] Open
Affiliation(s)
- Christian Hölscher
- Research and Experimental Center, Henan University of Chinese Medicine, 450046 Zhengzhou city, Henan province, PR China.
| |
Collapse
|