1
|
Espargaró A, Álvarez-Berbel I, Busquets MA, Sabate R. In Vivo Assays for Amyloid-Related Diseases. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2024; 17:433-458. [PMID: 38598824 DOI: 10.1146/annurev-anchem-061622-023326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Amyloid-related diseases, such as Alzheimer's and Parkinson's disease, are devastating conditions caused by the accumulation of abnormal protein aggregates known as amyloid fibrils. While assays involving animal models are essential for understanding the pathogenesis and developing therapies, a wide array of standard analytical techniques exists to enhance our understanding of these disorders. These techniques provide valuable information on the formation and propagation of amyloid fibrils, as well as the pharmacokinetics and pharmacodynamics of candidate drugs. Despite ethical concerns surrounding animal use, animal models remain vital tools in the search for treatments. Regardless of the specific animal model chosen, the analytical methods used are usually standardized. Therefore, the main objective of this review is to categorize and outline the primary analytical methods used in in vivo assays for amyloid-related diseases, highlighting their critical role in furthering our understanding of these disorders and developing effective therapies.
Collapse
Affiliation(s)
- Alba Espargaró
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
- 2Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Irene Álvarez-Berbel
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
| | - Maria Antònia Busquets
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
- 2Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Raimon Sabate
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
- 2Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| |
Collapse
|
2
|
Ono M, Ito T, Yamaki S, Hori Y, Zhou Q, Zhao X, Muramoto S, Yamamoto R, Furuyama T, Sakata-Haga H, Hatta T, Hamaguchi T, Kato N. Spatiotemporal development of the neuronal accumulation of amyloid precursor protein and the amyloid plaque formation in the brain of 3xTg-AD mice. Heliyon 2024; 10:e28821. [PMID: 38596059 PMCID: PMC11002285 DOI: 10.1016/j.heliyon.2024.e28821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 03/25/2024] [Accepted: 03/25/2024] [Indexed: 04/11/2024] Open
Abstract
The amyloid plaque is a hallmark of Alzheimer's disease. The accumulation of the amyloid precursor protein (APP) in the neuronal structure is assumed to lead to amyloid plaque formation through the excessive production of β-amyloid protein. To study the relationship between the neuronal accumulation of APP and amyloid plaque formation, we histologically analyzed their development in the different brain regions in 3xTg-AD mice, which express Swedish mutated APP (APPSWE) in the neurons. Observation throughout the brain revealed APPSWE-positive somata in the broad regions. Quantitative model analysis showed that the somatic accumulation of APPSWE developed firstly in the hippocampus from a very early age (<1 month) and proceeded slower in the isocortex. In line with this, the hippocampus was the first region to form amyloid plaques at the age of 9-12 months, while amyloid plaques were rarely observed in the isocortex. Females had more APPSWE-positive somata and plaques than males. Furthermore, amyloid plaques were observed in the lateral septum and pontine grey, which did not contain APPSWE-positive somata but only the APPSWE-positive fibers. These results suggested that neuronal accumulation of APPSWE, both in somatodendritic and axonal domains, is closely related to the formation of amyloid plaques.
Collapse
Affiliation(s)
- Munenori Ono
- Department of Physiology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Tetsufumi Ito
- Systems Function and Morphology, University of Toyama, Toyama, 930-0194, Japan
| | - Sachiko Yamaki
- Department of Physiology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Yoshie Hori
- Department of Physiology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Qing Zhou
- Department of Physiology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Xirun Zhao
- Department of Physiology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Shinji Muramoto
- Department of Physiology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Ryo Yamamoto
- Department of Physiology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Takafumi Furuyama
- Department of Physiology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Hiromi Sakata-Haga
- Department of Anatomy, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Toshihisa Hatta
- Department of Anatomy, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Tsuyoshi Hamaguchi
- Department of Neurology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Nobuo Kato
- Department of Physiology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| |
Collapse
|
3
|
Dean B, Duce J, Li QX, Masters CL, Scarr E. Lower levels of soluble β-amyloid precursor protein, but not β-amyloid, in the frontal cortex in schizophrenia. Psychiatry Res 2024; 331:115656. [PMID: 38071879 DOI: 10.1016/j.psychres.2023.115656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/28/2023] [Accepted: 12/01/2023] [Indexed: 01/02/2024]
Abstract
We identified a sub-group (25%) of people with schizophrenia (muscarinic receptor deficit schizophrenia (MRDS)) that are characterised because of markedly lower levels of cortical muscarinic M1 receptors (CHRM1) compared to most people with the disorder (non-MRDS). Notably, bioinformatic analyses of our cortical gene expression data shows a disturbance in the homeostasis of a biochemical pathway that regulates levels of CHRM1. A step in this pathway is the processing of β-amyloid precursor protein (APP) and therefore we postulated there would be altered levels of APP in the frontal cortex from people with MRDS. Here we measure levels of CHRM1 using [3H]pirenzepine binding, soluble APP (sAPP) using Western blotting and amyloid beta peptides (Aβ1-40 and Aβ1-42) using ELISA in the frontal cortex (Brodmann's area 6: BA 6; MRDS = 14, non-MRDS = 14, controls = 14). We confirmed the MRDS cohort in this study had the expected low levels of [3H]pirenzepine binding. In addition, we showed that people with schizophrenia, independent of their sub-group status, had lower levels of sAPP compared to controls but did not have altered levels of Aβ1-40 or Aβ1-42. In conclusion, whilst changes in sAPP are not restricted to MRDS our data could indicate a role of APP, which is important in axonal and synaptic pruning, in the molecular pathology of the syndrome of schizophrenia.
Collapse
Affiliation(s)
- Brian Dean
- The Florey, Parkville, Victoria, Australia; The University of Melbourne of Melbourne Florey Department of Neuroscience and Mental Health, Parkville, Victoria, Australia.
| | - James Duce
- MSD Discovery Centre, 120 Moorgate, London, UK
| | - Qiao-Xin Li
- The Florey, Parkville, Victoria, Australia; The University of Melbourne of Melbourne Florey Department of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Colin L Masters
- The Florey, Parkville, Victoria, Australia; The University of Melbourne of Melbourne Florey Department of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Elizabeth Scarr
- The Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
4
|
Huang Y, Zhang W, Guo X, Zhang Y, Wu J, Zu H. Cellular cholesterol loss by DHCR24 knockdown leads to Aβ production by changing APP intracellular localization. J Lipid Res 2023; 64:100367. [PMID: 37011864 PMCID: PMC10173783 DOI: 10.1016/j.jlr.2023.100367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/23/2023] [Accepted: 03/16/2023] [Indexed: 04/03/2023] Open
Abstract
For the past 20 years, the majority of cell culture studies reported that increasing cholesterol level increases amyloid-β (Aβ) production. Conversely, other studies and genetic evidences support that cellular cholesterol loss leads to Aβ generation. As a highly controversial issue in Alzheimer's disease (AD) pathogenesis,the apparent contradiction prompted us to again explore the role of cellular cholesterol in Aβ production. Here, we adopted new neuronal and astrocytic cell models induced by 3β-hydroxysterol-Δ24 reductase (DHCR24), which obviously differ from the widely used cell models with overexpressing amyloid precursor protein (APP) in the majority of previous studies. In neuronal and astrocytic cell model, we found that deficiency of cellular cholesterol by DHCR24 knock-down obviously increased intracellular and extracellular Aβ generation. Importantly, in cell models with overexpressing APP, we found that APP overexpression could disrupt cellular cholesterol homeostasis and affect function of cells, coupled with the increase of APP β-cleavage product, 99-residue transmembrane C-terminal domain (C99). Therefore, we suppose the results derived from the APP knock-in models will need to be re-evaluated. One rational explanation for the discrepancy between our outcomes and the previous studies could be attributed to the two different cell models. Mechanistically, we showed that cellular cholesterol loss obviously altered APP intracellular localization by affecting cholesterol-related trafficking protein of APP. Therefore, our outcomes strongly support cellular cholesterol loss by DHCR24 knockdown leads to Aβ production.
Collapse
|
5
|
Kobro-Flatmoen A, Battistin C, Nair RR, Bjorkli C, Skender B, Kentros C, Gouras G, Witter MP. Lowering levels of reelin in entorhinal cortex layer II-neurons results in lowered levels of intracellular amyloid-β. Brain Commun 2023; 5:fcad115. [PMID: 37091586 PMCID: PMC10120433 DOI: 10.1093/braincomms/fcad115] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 02/14/2023] [Accepted: 04/05/2023] [Indexed: 04/25/2023] Open
Abstract
Projection neurons in the anteriolateral part of entorhinal cortex layer II are the predominant cortical site for hyper-phosphorylation of tau and formation of neurofibrillary tangles in prodromal Alzheimer's disease. A majority of layer II projection neurons in anteriolateral entorhinal cortex are unique among cortical excitatory neurons by expressing the protein reelin. In prodromal Alzheimer's disease, these reelin-expressing neurons are prone to accumulate intracellular amyloid-β, which is mimicked in a rat model that replicates the spatio-temporal cascade of the disease. Two important findings in relation to this are that reelin-signalling downregulates tau phosphorylation, and that oligomeric amyloid-β interferes with reelin-signalling. Taking advantage of this rat model, we used proximity ligation assay to assess whether reelin and intracellular amyloid-β directly interact during early, pre-plaque stages in anteriolateral entorhinal cortex layer II reelin-expressing neurons. We next made a viral vector delivering micro-RNA against reelin, along with a control vector, and infected reelin-expressing anteriolateral entorhinal cortex layer II-neurons to test whether reelin levels affect levels of intracellular amyloid-β and/or amyloid precursor protein. We analysed 25.548 neurons from 24 animals, which results in three important findings. First, in reelin-expressing anteriolateral entorhinal cortex layer II-neurons, reelin and intracellular amyloid-β engage in a direct protein-protein interaction. Second, injecting micro-RNA against reelin lowers reelin levels in these neurons, amounting to an effect size of 1.3-4.5 (Bayesian estimation of Cohen's d effect size, 95% credible interval). This causes a concomitant reduction of intracellular amyloid-β ranging across three levels of aggregation, including a reduction of Aβ42 monomers/dimers amounting to an effect size of 0.5-3.1, a reduction of Aβ prefibrils amounting to an effect size of 1.1-3.5 and a reduction of protofibrils amounting to an effect size of 0.05-2.1. Analysing these data using Bayesian estimation of mutual information furthermore reveals that levels of amyloid-β are dependent on levels of reelin. Third, the reduction of intracellular amyloid-β occurs without any substantial associated changes in levels of amyloid precursor protein. We conclude that reelin and amyloid-β directly interact at the intracellular level in the uniquely reelin-expressing projection neurons in anteriolateral entorhinal cortex layer II, where levels of amyloid-β are dependent on levels of reelin. Since amyloid-β is known to impair reelin-signalling causing upregulated phosphorylation of tau, our findings are likely relevant to the vulnerability for neurofibrillary tangle-formation of this entorhinal neuronal population.
Collapse
Affiliation(s)
| | - Claudia Battistin
- Kavli Institute for Systems Neuroscience MTFS, NTNU Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7489, Trondheim, Norway
| | - Rajeevkumar Raveendran Nair
- Kavli Institute for Systems Neuroscience MTFS, NTNU Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7489, Trondheim, Norway
| | - Christiana Bjorkli
- Kavli Institute for Systems Neuroscience MTFS, NTNU Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7489, Trondheim, Norway
| | - Belma Skender
- Kavli Institute for Systems Neuroscience MTFS, NTNU Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7489, Trondheim, Norway
| | - Cliff Kentros
- Kavli Institute for Systems Neuroscience MTFS, NTNU Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7489, Trondheim, Norway
- Mohn Research Center for the Brain, NTNU, 7489, Trondheim, Norway
- Institute of Neuroscience, University of Oregon, 97401, Eugene, OR, USA
| | - Gunnar Gouras
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Menno P Witter
- Correspondence to: Menno P. Witter Kavli Institute for Systems Neuroscience MTFS, NTNU Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7489, Trondheim, Norway 7030 Trondheim, Norway E-mail:
| |
Collapse
|
6
|
Reactive astrocytes acquire neuroprotective as well as deleterious signatures in response to Tau and Aß pathology. Nat Commun 2022; 13:135. [PMID: 35013236 PMCID: PMC8748982 DOI: 10.1038/s41467-021-27702-w] [Citation(s) in RCA: 116] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 12/06/2021] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) alters astrocytes, but the effect of Aß and Tau pathology is poorly understood. TRAP-seq translatome analysis of astrocytes in APP/PS1 ß-amyloidopathy and MAPTP301S tauopathy mice revealed that only Aß influenced expression of AD risk genes, but both pathologies precociously induced age-dependent changes, and had distinct but overlapping signatures found in human post-mortem AD astrocytes. Both Aß and Tau pathology induced an astrocyte signature involving repression of bioenergetic and translation machinery, and induction of inflammation pathways plus protein degradation/proteostasis genes, the latter enriched in targets of inflammatory mediator Spi1 and stress-activated cytoprotective Nrf2. Astrocyte-specific Nrf2 expression induced a reactive phenotype which recapitulated elements of this proteostasis signature, reduced Aß deposition and phospho-tau accumulation in their respective models, and rescued brain-wide transcriptional deregulation, cellular pathology, neurodegeneration and behavioural/cognitive deficits. Thus, Aß and Tau induce overlapping astrocyte profiles associated with both deleterious and adaptive-protective signals, the latter of which can slow patho-progression.
Collapse
|
7
|
Villanueva EB, Tresse E, Liu Y, Duarte JN, Jimenez-Duran G, Ejlerskov P, Kretz O, Loreth D, Goldmann T, Prinz M, Issazadeh-Navikas S. Neuronal TNFα, Not α-Syn, Underlies PDD-Like Disease Progression in IFNβ-KO Mice. Ann Neurol 2021; 90:789-807. [PMID: 34476836 DOI: 10.1002/ana.26209] [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: 03/17/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Parkinson's disease (PD) manifests in motor dysfunction, non-motor symptoms, and eventual dementia (PDD). Neuropathological hallmarks include nigrostriatal neurodegeneration, Lewy body (LB) pathology, and neuroinflammation. Alpha-synuclein (α-syn), a primary component of LBs, is implicated in PD pathogenesis, accumulating, and aggregating in both familial and sporadic PD. However, as α-syn pathology is often comorbid with amyloid-beta (Aβ) plaques and phosphorylated tau (pTau) tangles in PDD, it is still unclear whether α-syn is the primary cause of neurodegeneration in sporadic PDD. We aimed to determine how the absence of α-syn would affect PDD manifestation. METHODS IFN-β knockout (Ifnb-/- ) mice spontaneously develop progressive behavior abnormalities and neuropathology resembling PDD, notably with α-syn+ LBs. We generated Ifnb/Snca double knockout (DKO) mice and evaluated their behavior and neuropathology compared with wild-type (Wt), Ifnb-/- , and Snca-/- mice using immunohistochemistry, electron microscopy, immunoblots, qPCR, and modification of neuronal signaling. RESULTS Ifnb/Snca DKO mice developed all clinical PDD-like behavioral manifestations induced by IFN-β loss. Independently of α-syn expression, lack of IFN-β alone induced Aβ plaques, pTau tangles, and LB-like Aβ+ /pTau+ inclusion bodies and neuroinflammation. IFN-β loss caused significant elevated glial and neuronal TNF-α and neuronal TNFR1, associated with neurodegeneration. Restoring neuronal IFN-β signaling or blocking TNFR1 rescued caspase 3/t-BID-mediated neuronal-death through upregulation of c-FLIPS and lowered intraneuronal Aβ and pTau accumulation. INTERPRETATION These findings increase our understanding of PD pathology and suggest that targeting α-syn alone is not sufficient to mitigate disease. Targeting specific aspects of neuroinflammation, such as aberrant neuronal TNF-α/TNFR1 or IFN-β/IFNAR signaling, may attenuate disease. ANN NEUROL 2021;90:789-807.
Collapse
Affiliation(s)
- Erika B Villanueva
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emilie Tresse
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yawei Liu
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - João N Duarte
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gisela Jimenez-Duran
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Patrick Ejlerskov
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Oliver Kretz
- Department of Internal Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Desiree Loreth
- Institute of Cellular and Integrative Physiology, University Clinic Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Goldmann
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiberg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiberg, Germany
| | - Shohreh Issazadeh-Navikas
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
Singh CSB, Choi KB, Munro L, Wang HY, Pfeifer CG, Jefferies WA. Reversing pathology in a preclinical model of Alzheimer's disease by hacking cerebrovascular neoangiogenesis with advanced cancer therapeutics. EBioMedicine 2021; 71:103503. [PMID: 34534764 PMCID: PMC8449085 DOI: 10.1016/j.ebiom.2021.103503] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/28/2021] [Accepted: 07/13/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Cognitive decline leading to dementia, accompanied by the accumulation of amyloid-beta (Aβ) in neuritic plaques together with the appearance of neurofibrillary tangles (NFT) composed of hyperphosphorylated tau protein (tau), are previously noted hallmarks of Alzheimer's disease (AD). We previously discovered hypervascularity in brain specimens from AD patients and consistent with this observation, we demonstrated that overexpression of Aβ drives cerebrovascular neoangiogenesis leading to hypervascularity and coincident tight-junction disruption and blood-brain barrier (BBB) leakiness in animal models of AD. We subsequently demonstrated that amyloid plaque burden and cerebrovascular pathogenesis subside when pro-angiogenic Aβ levels are reduced. Based on these data, we propose a paradigm of AD etiology where, as a compensatory response to impaired cerebral blood flow (CBF), Aβ triggers pathogenic cerebrovascular neoangiogenesis that underlies the conventional hallmarks of AD. Consequently, here we present evidence that repurposing anti-cancer drugs to modulate cerebrovascular neoangiogenesis, rather than directly targeting the amyloid cascade, may provide an effective treatment for AD and related vascular diseases of the brain. METHODS We explored whether the anti-cancer drug, Axitinib, a small molecule tyrosine kinase inhibitor that targets vascular endothelial growth factor receptors (VEGFR) can inhibit aberrant cerebrovascular neoangiogenic changes, reduce Aβ deposits and reverse cognitive decline in an animal model of AD. One month post-treatment with Axitinib, we employed a battery of tests to assess cognition and memory in aged Tg2576 AD mice and used molecular analysis to demonstrate reduction of amyloid plaques, BBB leakage, hypervascularity and associated disease pathology. FINDINGS Targeting the pro-angiogenic pathway in AD using the cancer drug, Axitinib, dramatically reduced cerebrovascular neoangiogenesis, restored BBB integrity, resolved tight-junction pathogenesis, diminishes Aβ depositions in plaques and effectively restores memory and cognitive performance in a preclinical mouse model of AD. INTERPRETATION Modulation of neoangiogenesis, in an analogous approach to those used to treat aberrant vascularization in cancer and also in the wet form of age-related macular degeneration (AMD), provides an alternative therapeutic strategy for intervention in AD that warrants clinical investigation. FUNDING None.
Collapse
Affiliation(s)
- Chaahat S B Singh
- Department of Medical Genetics, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC V6T 1Z4, Canada; Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z4, Canada; The Vancouver Prostate Centre, Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6T 1Z4, Canada
| | - Kyung Bok Choi
- Department of Medical Genetics, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC V6T 1Z4, Canada; Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z4, Canada; Department of Microbiology and Immunology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; Department of Zoology, University of British Columbia, 6270 University Blvd., Vancouver, BC V6T 1Z4, Canada; The Vancouver Prostate Centre, Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6T 1Z4, Canada
| | - Lonna Munro
- Department of Medical Genetics, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC V6T 1Z4, Canada; Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z4, Canada; Department of Microbiology and Immunology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; Department of Zoology, University of British Columbia, 6270 University Blvd., Vancouver, BC V6T 1Z4, Canada; The Vancouver Prostate Centre, Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6T 1Z4, Canada
| | - Hong Yue Wang
- Department of Medical Genetics, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC V6T 1Z4, Canada
| | - Cheryl G Pfeifer
- Department of Medical Genetics, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC V6T 1Z4, Canada; Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z4, Canada; Department of Microbiology and Immunology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; Department of Zoology, University of British Columbia, 6270 University Blvd., Vancouver, BC V6T 1Z4, Canada; The Vancouver Prostate Centre, Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6T 1Z4, Canada
| | - Wilfred A Jefferies
- Department of Medical Genetics, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC V6T 1Z4, Canada; Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z4, Canada; Department of Microbiology and Immunology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z4, Canada; Department of Zoology, University of British Columbia, 6270 University Blvd., Vancouver, BC V6T 1Z4, Canada; The Vancouver Prostate Centre, Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6T 1Z4, Canada; Department of Urologic Sciences, University of British Columbia, Gordon & Leslie Diamond Health Care Centre, Level 6, 2775 Laurel Street, Vancouver, BC V5Z 1M9, Canada.
| |
Collapse
|
9
|
Pagnon de la Vega M, Giedraitis V, Michno W, Kilander L, Güner G, Zielinski M, Löwenmark M, Brundin R, Danfors T, Söderberg L, Alafuzoff I, Nilsson LNG, Erlandsson A, Willbold D, Müller SA, Schröder GF, Hanrieder J, Lichtenthaler SF, Lannfelt L, Sehlin D, Ingelsson M. The Uppsala APP deletion causes early onset autosomal dominant Alzheimer's disease by altering APP processing and increasing amyloid β fibril formation. Sci Transl Med 2021; 13:13/606/eabc6184. [PMID: 34380771 DOI: 10.1126/scitranslmed.abc6184] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 02/05/2021] [Accepted: 06/22/2021] [Indexed: 12/23/2022]
Abstract
Point mutations in the amyloid precursor protein gene (APP) cause familial Alzheimer's disease (AD) by increasing generation or altering conformation of amyloid β (Aβ). Here, we describe the Uppsala APP mutation (Δ690-695), the first reported deletion causing autosomal dominant AD. Affected individuals have an age at symptom onset in their early forties and suffer from a rapidly progressing disease course. Symptoms and biomarkers are typical of AD, with the exception of normal cerebrospinal fluid (CSF) Aβ42 and only slightly pathological amyloid-positron emission tomography signals. Mass spectrometry and Western blot analyses of patient CSF and media from experimental cell cultures indicate that the Uppsala APP mutation alters APP processing by increasing β-secretase cleavage and affecting α-secretase cleavage. Furthermore, in vitro aggregation studies and analyses of patient brain tissue samples indicate that the longer form of mutated Aβ, AβUpp1-42Δ19-24, accelerates the formation of fibrils with unique polymorphs and their deposition into amyloid plaques in the affected brain.
Collapse
Affiliation(s)
- María Pagnon de la Vega
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, 75185 Uppsala, Sweden
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, 75185 Uppsala, Sweden
| | - Wojciech Michno
- Department of Psychiatry and Neurochemistry, University of Gothenburg, 43180 Gothenburg, Sweden.,Department of Neuroscience, Physiology and Pharmacology, University College London, WC1E 6BT London, UK
| | - Lena Kilander
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, 75185 Uppsala, Sweden
| | - Gökhan Güner
- German Center for Neurodegenerative Diseases (DZNE) and Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81377 Munich, Germany
| | - Mara Zielinski
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7) and JuStruct, Jülich Center for Structural Biology, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Malin Löwenmark
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, 75185 Uppsala, Sweden
| | - RoseMarie Brundin
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, 75185 Uppsala, Sweden
| | - Torsten Danfors
- Department of Surgical Sciences, Radiology, Uppsala University, 75185 Uppsala, Sweden
| | | | - Irina Alafuzoff
- Department of Immunology, Genetics and Pathology, Clinical and Experimental Pathology, Uppsala University, 75185 Uppsala, Sweden
| | - Lars N G Nilsson
- Department of Pharmacology, University of Oslo and Oslo University Hospital, 0316 Oslo, Norway
| | - Anna Erlandsson
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, 75185 Uppsala, Sweden
| | - Dieter Willbold
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7) and JuStruct, Jülich Center for Structural Biology, Forschungszentrum Jülich, 52425 Jülich, Germany.,Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany.,Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, State University, 141701 Dolgoprudny, Russia
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE) and Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81377 Munich, Germany
| | - Gunnar F Schröder
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7) and JuStruct, Jülich Center for Structural Biology, Forschungszentrum Jülich, 52425 Jülich, Germany.,Physics Department, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, University of Gothenburg, 43180 Gothenburg, Sweden.,Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE) and Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81377 Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Lars Lannfelt
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, 75185 Uppsala, Sweden
| | - Dag Sehlin
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, 75185 Uppsala, Sweden
| | - Martin Ingelsson
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, 75185 Uppsala, Sweden.
| |
Collapse
|
10
|
Rice HC, Marcassa G, Chrysidou I, Horré K, Young-Pearse TL, Müller UC, Saito T, Saido TC, Vassar R, de Wit J, De Strooper B. Contribution of GABAergic interneurons to amyloid-β plaque pathology in an APP knock-in mouse model. Mol Neurodegener 2020; 15:3. [PMID: 31915042 PMCID: PMC6950898 DOI: 10.1186/s13024-019-0356-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 12/30/2019] [Indexed: 12/04/2022] Open
Abstract
The amyloid-β (Aβ) peptide, the primary constituent of amyloid plaques found in Alzheimer's disease (AD) brains, is derived from sequential proteolytic processing of the Amyloid Precursor Protein (APP). However, the contribution of different cell types to Aβ deposition has not yet been examined in an in vivo, non-overexpression system. Here, we show that endogenous APP is highly expressed in a heterogeneous subset of GABAergic interneurons throughout various laminae of the hippocampus, suggesting that these cells may have a profound contribution to AD plaque pathology. We then characterized the laminar distribution of amyloid burden in the hippocampus of an APP knock-in mouse model of AD. To examine the contribution of GABAergic interneurons to plaque pathology, we blocked Aβ production specifically in these cells using a cell type-specific knock-out of BACE1. We found that during early stages of plaque deposition, interneurons contribute to approximately 30% of the total plaque load in the hippocampus. The greatest contribution to plaque load (75%) occurs in the stratum pyramidale of CA1, where plaques in human AD cases are most prevalent and where pyramidal cell bodies and synaptic boutons from perisomatic-targeting interneurons are located. These findings reveal a crucial role of GABAergic interneurons in the pathology of AD. Our study also highlights the necessity of using APP knock-in models to correctly evaluate the cellular contribution to amyloid burden since APP overexpressing transgenic models drive expression in cell types according to the promoter and integration site and not according to physiologically relevant expression mechanisms.
Collapse
Affiliation(s)
- Heather C. Rice
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Present Address: Department of Biochemistry and Molecular Biology, Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Gabriele Marcassa
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Iordana Chrysidou
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Katrien Horré
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Tracy L. Young-Pearse
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Ulrike C. Müller
- Department of Functional Genomics, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
| | - 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, Nagoya, Japan
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Robert Vassar
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, USA
| | - Joris de Wit
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Bart De Strooper
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- UK-Dementia Research Institute at University College London, London, UK
| |
Collapse
|
11
|
Yates SC, Groeneboom NE, Coello C, Lichtenthaler SF, Kuhn PH, Demuth HU, Hartlage-Rübsamen M, Roßner S, Leergaard T, Kreshuk A, Puchades MA, Bjaalie JG. QUINT: Workflow for Quantification and Spatial Analysis of Features in Histological Images From Rodent Brain. Front Neuroinform 2019; 13:75. [PMID: 31849633 PMCID: PMC6901597 DOI: 10.3389/fninf.2019.00075] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 11/15/2019] [Indexed: 01/22/2023] Open
Abstract
Transgenic animal models are invaluable research tools for elucidating the pathways and mechanisms involved in the development of neurodegenerative diseases. Mechanistic clues can be revealed by applying labelling techniques such as immunohistochemistry or in situ hybridisation to brain tissue sections. Precision in both assigning anatomical location to the sections and quantifying labelled features is crucial for output validity, with a stereological approach or image-based feature extraction typically used. However, both approaches are restricted by the need to manually delineate anatomical regions. To circumvent this limitation, we present the QUINT workflow for quantification and spatial analysis of labelling in series of rodent brain section images based on available 3D reference atlases. The workflow is semi-automated, combining three open source software that can be operated without scripting knowledge, making it accessible to most researchers. As an example, a brain region-specific quantification of amyloid plaques across whole transgenic Tg2576 mouse brain series, immunohistochemically labelled for three amyloid-related antigens is demonstrated. First, the whole brain image series were registered to the Allen Mouse Brain Atlas to produce customised atlas maps adapted to match the cutting plan and proportions of the sections (QuickNII software). Second, the labelling was segmented from the original images by the Random Forest Algorithm for supervised classification (ilastik software). Finally, the segmented images and atlas maps were used to generate plaque quantifications for each region in the reference atlas (Nutil software). The method yielded comparable results to manual delineations and to the output of a stereological method. While the use case demonstrates the QUINT workflow for quantification of amyloid plaques only, the workflow is suited to all mouse or rat brain series with labelling that is visually distinct from the background, for example for the quantification of cells or labelled proteins.
Collapse
Affiliation(s)
- Sharon C Yates
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Nicolaas E Groeneboom
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Christopher Coello
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, and Institute for Advanced Study, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Peer-Hendrik Kuhn
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Hans-Ulrich Demuth
- Department of Molecular Drug Design and Target Validation Fraunhofer Institute for Cell Therapy and Immunology, Halle (Saale), Leipzig, Germany
| | | | - Steffen Roßner
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Trygve Leergaard
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Anna Kreshuk
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Maja A Puchades
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jan G Bjaalie
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
12
|
Joseph DJ, Liu C, Peng J, Liang G, Wei H. Isoflurane mediated neuropathological and cognitive impairments in the triple transgenic Alzheimer's mouse model are associated with hippocampal synaptic deficits in an age-dependent manner. PLoS One 2019; 14:e0223509. [PMID: 31600350 PMCID: PMC6786564 DOI: 10.1371/journal.pone.0223509] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/22/2019] [Indexed: 12/17/2022] Open
Abstract
Many in vivo studies suggest that inhalational anesthetics can accelerate or prevent the progression of neuropathology and cognitive impairments in Alzheimer Disease (AD), but the synaptic mechanisms mediating these ambiguous effects are unclear. Here, we show that repeated exposures of neonatal and old triple transgenic AD (3xTg) and non-transgenic (NonTg) mice to isoflurane (Iso) distinctly increased neurodegeneration as measured by S100β levels, intracellular Aβ, Tau oligomerization, and apoptotic markers. Spatial cognition measured by reference and working memory testing in the Morris Water Maze (MWM) were altered in young NonTg and 3xTg. Field recordings in the cornu ammonis 1 (CA1) hippocampus showed that neonatal control 3xTg mice exhibited hypo-excitable synaptic transmission, reduced paired-pulse facilitation (PPF), and normal long-term potentiation (LTP) compared to NonTg controls. By contrast, the old control 3xTg mice exhibited hyper-excitable synaptic transmission, enhanced PPF, and unstable LTP compared to NonTg controls. Repeated Iso exposures reduced synaptic transmission and PPF in neonatal NonTg and old 3xTg mice. LTP was normalized in old 3xTg mice, but reduced in neonates. By contrast, LTP was reduced in old but not neonatal NonTg mice. Our results indicate that Iso-mediated neuropathologic and cognitive defects in AD mice are associated with synaptic pathologies in an age-dependent manner. Based on these findings, the extent of this association with age and, possibly, treatment paradigms warrant further study.
Collapse
Affiliation(s)
- Donald J. Joseph
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Chunxia Liu
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China
| | - Jun Peng
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- Department of Anesthesiology, sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ge Liang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Huafeng Wei
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- * E-mail:
| |
Collapse
|
13
|
Höfling C, Shehabi E, Kuhn PH, Lichtenthaler SF, Hartlage-Rübsamen M, Roßner S. Cell Type-Specific Human APP Transgene Expression by Hippocampal Interneurons in the Tg2576 Mouse Model of Alzheimer's Disease. Front Neurosci 2019; 13:137. [PMID: 30853883 PMCID: PMC6395433 DOI: 10.3389/fnins.2019.00137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/06/2019] [Indexed: 01/21/2023] Open
Abstract
Amyloid precursor protein (APP) transgenic animal models of Alzheimer’s disease have become versatile tools for basic and translational research. However, there is great heterogeneity of histological, biochemical, and functional data between transgenic mouse lines, which might be due to different transgene expression patterns. Here, the expression of human APP (hAPP) by GABAergic hippocampal interneurons immunoreactive for the calcium binding proteins parvalbumin, calbindin, calretinin, and for the peptide hormone somatostatin was analyzed in Tg2576 mice by double immunofluorescent microscopy. Overall, there was no GABAergic interneuron subpopulation that did not express the transgene. On the other hand, in no case all neurons of such a subpopulation expressed hAPP. In dentate gyrus molecular layer and in stratum lacunosum moleculare less than 10% of hAPP-positive interneurons co-express any of these interneuron markers, whereas in stratum oriens hAPP-expressing neurons frequently co-express these interneuron markers to different proportions. We conclude that these neurons differentially contribute to deficits in young Tg2576 mice before the onset of Abeta plaque pathology. The detailed analysis of distinct brain region and neuron type-specific APP transgene expression patterns is indispensable to understand particular pathological features and mouse line-specific differences in neuronal and systemic functions.
Collapse
Affiliation(s)
- Corinna Höfling
- Paul-Flechsig-Institute for Brain Research, Leipzig University, Leipzig, Germany
| | - Emira Shehabi
- Paul-Flechsig-Institute for Brain Research, Leipzig University, Leipzig, Germany
| | - Peer-Hendrik Kuhn
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Stefan F Lichtenthaler
- Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.,Institute for Advanced Study, Technical University of Munich, Garching, Germany
| | | | - Steffen Roßner
- Paul-Flechsig-Institute for Brain Research, Leipzig University, Leipzig, Germany
| |
Collapse
|
14
|
Whitesell JD, Buckley AR, Knox JE, Kuan L, Graddis N, Pelos A, Mukora A, Wakeman W, Bohn P, Ho A, Hirokawa KE, Harris JA. Whole brain imaging reveals distinct spatial patterns of amyloid beta deposition in three mouse models of Alzheimer's disease. J Comp Neurol 2018; 527:2122-2145. [PMID: 30311654 DOI: 10.1002/cne.24555] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 09/13/2018] [Indexed: 01/08/2023]
Abstract
A variety of Alzheimer's disease (AD) mouse models overexpress mutant forms of human amyloid precursor protein (APP), producing high levels of amyloid β (Aβ) and forming plaques. However, the degree to which these models mimic spatiotemporal patterns of Aβ deposition in brains of AD patients is unknown. Here, we mapped the spatial distribution of Aβ plaques across age in three APP-overexpression mouse lines (APP/PS1, Tg2576, and hAPP-J20) using in vivo labeling with methoxy-X04, high throughput whole brain imaging, and an automated informatics pipeline. Images were acquired with high resolution serial two-photon tomography and labeled plaques were detected using custom-built segmentation algorithms. Image series were registered to the Allen Mouse Brain Common Coordinate Framework, a 3D reference atlas, enabling automated brain-wide quantification of plaque density, number, and location. In both APP/PS1 and Tg2576 mice, plaques were identified first in isocortex, followed by olfactory, hippocampal, and cortical subplate areas. In hAPP-J20 mice, plaque density was highest in hippocampal areas, followed by isocortex, with little to no involvement of olfactory or cortical subplate areas. Within the major brain divisions, distinct regions were identified with high (or low) plaque accumulation; for example, the lateral visual area within the isocortex of APP/PS1 mice had relatively higher plaque density compared with other cortical areas, while in hAPP-J20 mice, plaques were densest in the ventral retrosplenial cortex. In summary, we show how whole brain imaging of amyloid pathology in mice reveals the extent to which a given model recapitulates the regional Aβ deposition patterns described in AD.
Collapse
Affiliation(s)
| | | | - Joseph E Knox
- Allen Institute for Brain Science, Seattle, Washington
| | - Leonard Kuan
- Allen Institute for Brain Science, Seattle, Washington
| | - Nile Graddis
- Allen Institute for Brain Science, Seattle, Washington
| | - Andrew Pelos
- Allen Institute for Brain Science, Seattle, Washington.,Department of Neuroscience, Pomona College, Claremont, California
| | - Alice Mukora
- Allen Institute for Brain Science, Seattle, Washington
| | - Wayne Wakeman
- Allen Institute for Brain Science, Seattle, Washington
| | - Phillip Bohn
- Allen Institute for Brain Science, Seattle, Washington
| | - Anh Ho
- Allen Institute for Brain Science, Seattle, Washington
| | | | | |
Collapse
|
15
|
Heiland T, Zeitschel U, Puchades MA, Kuhn PH, Lichtenthaler SF, Bjaalie JG, Hartlage-Rübsamen M, Roßner S, Höfling C. Defined astrocytic expression of human amyloid precursor protein in Tg2576 mouse brain. Glia 2018; 67:393-403. [PMID: 30485540 PMCID: PMC6588085 DOI: 10.1002/glia.23550] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/25/2018] [Accepted: 09/28/2018] [Indexed: 12/22/2022]
Abstract
Transgenic Tg2576 mice expressing human amyloid precursor protein (hAPP) with the Swedish mutation are among the most frequently used animal models to study the amyloid pathology related to Alzheimer's disease (AD). The transgene expression in this model is considered to be neuron‐specific. Using a novel hAPP‐specific antibody in combination with cell type‐specific markers for double immunofluorescent labelings and laser scanning microscopy, we here report that—in addition to neurons throughout the brain—astrocytes in the corpus callosum and to a lesser extent in neocortex express hAPP. This astrocytic hAPP expression is already detectable in young Tg2576 mice before the onset of amyloid pathology and still present in aged Tg2576 mice with robust amyloid pathology in neocortex, hippocampus, and corpus callosum. Surprisingly, hAPP immunoreactivity in cortex is restricted to resting astrocytes distant from amyloid plaques but absent from reactive astrocytes in close proximity to amyloid plaques. In contrast, neither microglial cells nor oligodendrocytes of young or aged Tg2576 mice display hAPP labeling. The astrocytic expression of hAPP is substantiated by the analyses of hAPP mRNA and protein expression in primary cultures derived from Tg2576 offspring. We conclude that astrocytes, in particular in corpus callosum, may contribute to amyloid pathology in Tg2576 mice and thus mimic this aspect of AD pathology.
Collapse
Affiliation(s)
- Tina Heiland
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Ulrike Zeitschel
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Maja A Puchades
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Peer-Hendrik Kuhn
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Stefan F Lichtenthaler
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Munich, Germany.,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Institute for Advanced Study, Technical University of Munich, Garching, Germany
| | - Jan G Bjaalie
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | - Steffen Roßner
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Corinna Höfling
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| |
Collapse
|
16
|
Ourdev D, Schmaus A, Kar S. Kainate Receptor Activation Enhances Amyloidogenic Processing of APP in Astrocytes. Mol Neurobiol 2018; 56:5095-5110. [PMID: 30484111 DOI: 10.1007/s12035-018-1427-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/13/2018] [Indexed: 12/21/2022]
Abstract
Kainic acid (KA) is an analogue of the excitatory neurotransmitter glutamate that, when injected systemically into adult rats, can trigger seizures and progressive neuronal loss in a manner that mirrors the neuropathology of human mesial temporal lobe epilepsy. However, biomolecular mechanisms responsible for the neuronal loss that occurs as a consequence of this treatment remains elusive. We have recently reported that toxicity induced by KA can partly be mediated by astrocyte-derived amyloid β (Aβ) peptides, which are critical in the development of Alzheimer's disease (AD). Nonetheless, little is known how KA can influence amyloid precursor protein (APP) levels and processing in astrocytes. Thus, in the present study using human U-373 astrocytoma and rat primary astrocytes, we evaluated the role of KA on APP metabolism. Our results revealed that KA treatment increased the levels of APP and its cleaved products (α-/β-CTFs) in cultured U-373 astrocytoma and primary astrocytes, without altering the cell viability. The cellular and secretory levels of Aβ1-40/Aβ1-42 were markedly increased in KA-treated astrocytes. We also demonstrated that the steady-state levels of APP-secretases were not altered but the activity of γ-secretase is enhanced in KA-treated U-373 astrocytoma. Furthermore, using selective receptor antagonists, we showed that the effects of KA is mediated by activation of kainate receptors and not NMDA or AMPA receptors. These results suggest that KA can enhance amyloidogenic processing of APP by activating its own receptor leading to increased production/secretion of Aβ-related peptides from activated astrocytes which may contribute to the pathogenesis of temporal lobe epilepsy.
Collapse
Affiliation(s)
- D Ourdev
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada.,Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
| | - A Schmaus
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Satyabrata Kar
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada. .,Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada. .,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada. .,Department of Medicine, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
17
|
Endosomal-Lysosomal Cholesterol Sequestration by U18666A Differentially Regulates Amyloid Precursor Protein (APP) Metabolism in Normal and APP-Overexpressing Cells. Mol Cell Biol 2018. [PMID: 29530923 DOI: 10.1128/mcb.00529-17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Amyloid β (Aβ) peptide, derived from amyloid precursor protein (APP), plays a critical role in the development of Alzheimer's disease. Current evidence indicates that altered levels or subcellular distribution of cholesterol can regulate Aβ production and clearance, but it remains unclear how cholesterol sequestration within the endosomal-lysosomal (EL) system can influence APP metabolism. Thus, we evaluated the effects of U18666A, which triggers cholesterol redistribution within the EL system, on mouse N2a cells expressing different levels of APP in the presence or absence of extracellular cholesterol and lipids provided by fetal bovine serum (FBS). Our results reveal that U18666A and FBS differentially increase the levels of APP and its cleaved products, the α-, β-, and η-C-terminal fragments, in N2a cells expressing normal levels of mouse APP (N2awt), higher levels of human wild-type APP (APPwt), or "Swedish" mutant APP (APPsw). The cellular levels of Aβ1-40/Aβ1-42 were markedly increased in U18666A-treated APPwt and APPsw cells. Our studies further demonstrate that APP and its cleaved products are partly accumulated in the lysosomes, possibly due to decreased clearance. Finally, we show that autophagy inhibition plays a role in mediating U18666A effects. Collectively, these results suggest that altered levels and distribution of cholesterol and lipids can differentially regulate APP metabolism depending on the nature of APP expression.
Collapse
|
18
|
Hartlage-Rübsamen M, Bluhm A, Piechotta A, Linnert M, Rahfeld JU, Demuth HU, Lues I, Kuhn PH, Lichtenthaler SF, Roßner S, Höfling C. Immunohistochemical Evidence from APP-Transgenic Mice for Glutaminyl Cyclase as Drug Target to Diminish pE-Abeta Formation. Molecules 2018; 23:molecules23040924. [PMID: 29673150 PMCID: PMC6017857 DOI: 10.3390/molecules23040924] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/05/2018] [Accepted: 04/10/2018] [Indexed: 02/06/2023] Open
Abstract
Oligomeric assemblies of neurotoxic amyloid beta (Abeta) peptides generated by proteolytical processing of the amyloid precursor protein (APP) play a key role in the pathogenesis of Alzheimer’s disease (AD). In recent years, a substantial heterogeneity of Abeta peptides with distinct biophysical and cell biological properties has been demonstrated. Among these, a particularly neurotoxic and disease-specific Abeta variant is N-terminally truncated and modified to pyroglutamate (pE-Abeta). Cell biological and animal experimental studies imply the catalysis of this modification by the enzyme glutaminyl cyclase (QC). However, direct histopathological evidence in transgenic animals from comparative brain region and cell type-specific expression of transgenic hAPP and QC, on the one hand, and on the formation of pE-Abeta aggregates, on the other, is lacking. Here, using single light microscopic, as well as triple immunofluorescent, labeling, we report the deposition of pE-Abeta only in the brain regions of APP-transgenic Tg2576 mice with detectable human APP and endogenous QC expression, such as the hippocampus, piriform cortex, and amygdala. Brain regions showing human APP expression without the concomitant presence of QC (the anterodorsal thalamic nucleus and perifornical nucleus) do not display pE-Abeta plaque formation. However, we also identified brain regions with substantial expression of human APP and QC in the absence of pE-Abeta deposition (the Edinger-Westphal nucleus and locus coeruleus). In these brain regions, the enzymes required to generate N-truncated Abeta peptides as substrates for QC might be lacking. Our observations provide additional evidence for an involvement of QC in AD pathogenesis via QC-catalyzed pE-Abeta formation.
Collapse
Affiliation(s)
| | - Alexandra Bluhm
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany.
| | - Anke Piechotta
- Department of Molecular Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, 06120 Halle (Saale), Germany.
| | - Miriam Linnert
- Department of Molecular Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, 06120 Halle (Saale), Germany.
| | - Jens-Ulrich Rahfeld
- Department of Molecular Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, 06120 Halle (Saale), Germany.
| | - Hans-Ulrich Demuth
- Department of Molecular Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, 06120 Halle (Saale), Germany.
| | - Inge Lues
- Probiodrug AG, 06120 Halle (Saale), Germany.
| | - Peer-Hendrik Kuhn
- Institute of Pathology, Technical University of Munich, 81675 Munich, Germany.
| | - Stefan F Lichtenthaler
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 81377 Munich, Germany.
- Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany.
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany.
- Institute for Advanced Study, Technical University of Munich, 85748 Garching, Germany.
| | - Steffen Roßner
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany.
| | - Corinna Höfling
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany.
| |
Collapse
|
19
|
Sasaguri H, Nilsson P, Hashimoto S, Nagata K, Saito T, De Strooper B, Hardy J, Vassar R, Winblad B, Saido TC. APP mouse models for Alzheimer's disease preclinical studies. EMBO J 2017; 36:2473-2487. [PMID: 28768718 PMCID: PMC5579350 DOI: 10.15252/embj.201797397] [Citation(s) in RCA: 483] [Impact Index Per Article: 60.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 06/09/2017] [Accepted: 07/07/2017] [Indexed: 12/11/2022] Open
Abstract
Animal models of human diseases that accurately recapitulate clinical pathology are indispensable for understanding molecular mechanisms and advancing preclinical studies. The Alzheimer's disease (AD) research community has historically used first‐generation transgenic (Tg) mouse models that overexpress proteins linked to familial AD (FAD), mutant amyloid precursor protein (APP), or APP and presenilin (PS). These mice exhibit AD pathology, but the overexpression paradigm may cause additional phenotypes unrelated to AD. Second‐generation mouse models contain humanized sequences and clinical mutations in the endogenous mouse App gene. These mice show Aβ accumulation without phenotypes related to overexpression but are not yet a clinical recapitulation of human AD. In this review, we evaluate different APP mouse models of AD, and review recent studies using the second‐generation mice. We advise AD researchers to consider the comparative strengths and limitations of each model against the scientific and therapeutic goal of a prospective preclinical study.
Collapse
Affiliation(s)
- Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako, Japan .,Department of Neurology and Neurological Science, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Per Nilsson
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako, Japan.,Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Huddinge, Sweden
| | - Shoko Hashimoto
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako, Japan
| | - Kenichi Nagata
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako, Japan
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako, Japan.,Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Bart De Strooper
- Dementia Research Institute, University College London, London, UK.,Department for Neurosciences, KU Leuven, Leuven, Belgium.,VIB Center for Brain and Disease Research, Leuven, Belgium
| | - John Hardy
- Reta Lila Research Laboratories and the Department of Molecular Neuroscience, University College London Institute of Neurology, London, UK
| | - Robert Vassar
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Huddinge, Sweden
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako, Japan
| |
Collapse
|