1
|
Kunze LH, Palumbo G, Gnörich J, Wind-Mark K, Schaefer R, Lindner S, Gildehaus FJ, Ziegler S, Brendel M. Fibrillar amyloidosis and synaptic vesicle protein expression progress jointly in the cortex of a mouse model with β-amyloid pathology. Neuroimage 2025; 310:121165. [PMID: 40120783 DOI: 10.1016/j.neuroimage.2025.121165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/13/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025] Open
Abstract
Neurodegeneration, accumulation of β-amyloid (Aβ) plaques, and neuroinflammation are the major hallmarks of Alzheimer's disease. Here, we aimed to investigate the temporal and spatial association between synaptic activity, Aβ plaque load, and neuroinflammation in an Aβ mouse model with limited neurodegeneration. 26 APPSL70 and 15 C57Bl/6 mice underwent longitudinal PET-scans with [18F]UCB-H from plaque onset to levels of strong plaque load (5.3 - 11.0 months of age) to assess the synaptic vesicle protein 2A (SV2A) expression, [18F]FBB to determine the fibrillar Aβ plaque load, and [18F]GE-180 and [18F]F-DED to assess microglial and astroglial (re)activity. Statistical parametric mapping was performed to uncover similarities between the binding patterns of all four tracers. We found a continuous increase in Aβ-PET in APPSL70 mice from 5.3 to 11.0 months of age, resulting in a significantly higher [18F]FBB PET signal in the cortex, hippocampus, and thalamus of APPSL70 mice compared to C57Bl/6 mice at 11.0 months of age. Parallel increases in SV2A-PET signals were observed in the cortex and thalamus of APPSL70 mice compared to C57Bl/6 mice. Statistical parametric mapping revealed a similar pattern of Aβ- and SV2A-PET differences (dice coefficient 53 %). Patterns of microglia activation showed stronger congruency with SV2A expression (dice coefficient 58 %) than patterns of reactive astrogliosis (dice coefficient 26 %). APPSL70 mice with limited neurodegeneration comprise a close temporal and spatial association between SV2A expression, Aβ plaque load, and microglial activation. SV2A PET imaging in APPSL70 mice may facilitate longitudinal monitoring of increased synaptic activity in the earliest phase of AD.
Collapse
Affiliation(s)
- L H Kunze
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - G Palumbo
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - J Gnörich
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - K Wind-Mark
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - R Schaefer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - S Lindner
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - F-J Gildehaus
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - S Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; Department of Nuclear Medicine, Hannover Medical School (MHH), Hannover, Germany
| | - M Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Ludwig Maximilian University of Munich, 81377 Munich, Germany.
| |
Collapse
|
2
|
Shojaei M, Schaefer R, Schlepckow K, Kunze LH, Struebing FL, Brunner B, Willem M, Bartos LM, Feiten A, Palumbo G, Arzberger T, Bartenstein P, Parico GC, Xia D, Monroe KM, Haass C, Brendel M, Lindner S. PET imaging of microglia in Alzheimer's disease using copper-64 labeled TREM2 antibodies. Theranostics 2024; 14:6319-6336. [PMID: 39431020 PMCID: PMC11488106 DOI: 10.7150/thno.97149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/11/2024] [Indexed: 10/22/2024] Open
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM2) plays an essential role in microglia activation and is being investigated as a potential therapeutic target for modulation of microglia in several neurological diseases. In this study, we present the development and preclinical evaluation of 64Cu-labeled antibody-based PET radiotracers as tools for non-invasive assessment of TREM2 expression. Furthermore, we tested the potential of an antibody transport vehicle (ATV) that binds human transferrin receptor to facilitate transcytosis of TREM2 antibody-based radiotracers to the CNS and improve target engagement. Methods: A TREM2 antibody with an engineered transport vehicle (ATV:4D9) and without (4D9) were covalently modified with pNCS-benzyl-NODAGA and labeled with copper-64. Potency, stability, and specificity were assessed in vitro followed by in vivo PET imaging at the early 2 h, intermediate 20 h, and late imaging time points 40 h post-injection using a human transferrin receptor (hTfR) expressing model for amyloidogenesis (5xFAD;TfRmu/hu) or wild-type mice (WT;TfRmu/hu), and hTfR negative controls. Organs of interest were isolated to determine biodistribution by ex vivo autoradiography. Cell sorting after in vivo tracer injection was used to demonstrate cellular specificity for microglia and to validate TREM2 PET results in an independent mouse model for amyloidogenesis (AppSAA;TfRmu/hu). For translation to human imaging, a human TREM2 antibody (14D3) was radiolabeled and used for in vitro autoradiography on human brain sections. Results: The 64Cu-labeled antibodies were obtained in high radiochemical purity (RCP), radiochemical yield (RCY), and specific activity. Antibody modification did not impact TREM2 binding. ATV:4D9 binding proved to be specific, and the tracer stability was maintained over 48 h. The uptake of [64Cu]Cu-NODAGA-ATV:4D9 in the brains of hTfR expressing mice was up to 4.6-fold higher than [64Cu]Cu-NODAGA-4D9 in mice without hTfR. TREM2 PET revealed elevated uptake in the cortex of 5xFAD mice compared to wild-type, which was validated by autoradiography. PET-to-biodistribution correlation revealed that elevated radiotracer uptake in brains of 5xFAD;TfRmu/hu mice was driven by microglia-rich cortical and hippocampal brain regions. Radiolabeled ATV:4D9 was selectively enriched in microglia and cellular uptake explained PET signal enhancement in AppSAA;TfRmu/hu mice. Human autoradiography showed elevated TREM2 tracer binding in the cortex of patients with Alzheimer's disease. Conclusion: [64Cu]Cu-NODAGA-ATV:4D9 has potential for non-invasive assessment of TREM2 as a surrogate marker for microglia activation in vivo. ATV engineering for hTfR binding and transcytosis overcomes the blood-brain barrier restriction for antibody-based PET radiotracers. TREM2 PET might be a versatile tool for many applications beyond Alzheimer's disease, such as glioma and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Monireh Shojaei
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Rebecca Schaefer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Kai Schlepckow
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Lea H. Kunze
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Felix L. Struebing
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Center for Neuropathology and Prion Research, University Hospital, LMU Munich, Munich, Germany
| | - Bettina Brunner
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Michael Willem
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Laura M. Bartos
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Astrid Feiten
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Munich, Germany
| | - Giovanna Palumbo
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Thomas Arzberger
- Center for Neuropathology and Prion Research, University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | | | - Dan Xia
- Denali Therapeutics Inc, South San Francisco, CA, USA
| | | | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
3
|
Lana D, Traini C, Bulli I, Sarti G, Magni G, Attorre S, Giovannini MG, Vannucchi MG. Chronic administration of prebiotics and probiotics ameliorates pathophysiological hallmarks of Alzheimer's disease in a APP/PS1 transgenic mouse model. Front Pharmacol 2024; 15:1451114. [PMID: 39166107 PMCID: PMC11333230 DOI: 10.3389/fphar.2024.1451114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
Introduction: The gut microbiota (MB), although one of the main producers of Aβ in the body, in physiological conditions contributes to the maintainance of a healthy brain. Dysbiosis, the dysbalance between Gram-negative and Gram-positive bacteria in the MB increases Aβ production, contributing to the accumulation of Aβ plaques in the brain, the main histopathological hallmark of Alzheimer's disease (AD). Administration of prebiotics and probiotics, maintaining or recovering gut-MB composition, could represent a nutraceutical strategy to prevent or reduce AD sympthomathology. Aim of this research was to evaluate whether treatment with pre- and probiotics could modify the histopathological signs of neurodegeneration in hippocampal CA1 and CA3 areas of a transgenic mouse model of AD (APP/PS1 mice). The hippocampus is one of the brain regions involved in AD. Methods: Tg mice and Wt littermates (Wt-T and Tg-T) were fed daily for 6 months from 2 months of age with a diet supplemented with prebiotics (a multi-extract of fibers and plant complexes, containing inulin/fruit-oligosaccharides) and probiotics (a 50%-50% mixture of Lactobacillus rhamnosus and Lactobacillus paracasei). Controls were Wt and Tg mice fed with a standard diet. Brain sections were immunostained for Aβ plaques, neurons, astrocytes, microglia, and inflammatory proteins that were evaluated qualitatively and quantitatively by immunofluorescence, confocal microscopy and digital imaging with ImageJ software. Results: Quantitative analyses demonstrated that: 1) The treatment with pre- and probiotics significantly decreased Aβ plaques in CA3, while in CA1 the reduction was not significant; 2) Neuronal damage in CA1 Stratum Pyramidalis was significantly prevented in Tg-T mice; no damage was found in CA3; 3) In both CA1 and CA3 the treatment significantly increased astrocytes density, and GFAP and IBA1 expression, especially around plaques; 4) Microglia reacted differently in CA1 and CA3: in CA3 of Tg-T mice there was a significant increase of CD68+ phagocytic microglia (ball-and-chain phenomic) and of CX3CR1 compared with CA1. Discussion: The higher microglia reactivity could be responsible for their more efficient scavenging activity towards Aβ plaques in CA3 in comparison to CA1. Treatment with pre- and probiotics, modifying many of the physiopathological hallmarks of AD, could be considered an effective nutraceutical strategy against AD symptomatology.
Collapse
Affiliation(s)
- Daniele Lana
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Chiara Traini
- Research Unit of Histology and Embryology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Irene Bulli
- Research Unit of Histology and Embryology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giorgia Sarti
- Research Unit of Histology and Embryology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giada Magni
- Cnr — Istituto di Fisica Applicata “Nello Carrara”, Sesto Fiorentino, Italy
| | - Selene Attorre
- Section of Anatomic Pathology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Maria Grazia Giovannini
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Maria Giuliana Vannucchi
- Research Unit of Histology and Embryology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
4
|
Ma X, Xing Y, Zhai R, Du Y, Yan H. Development and advancements in rodent MRI-based brain atlases. Heliyon 2024; 10:e27421. [PMID: 38510053 PMCID: PMC10950579 DOI: 10.1016/j.heliyon.2024.e27421] [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: 01/25/2023] [Revised: 02/15/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
Rodents, particularly mice and rats, are extensively utilized in fundamental neuroscience research. Brain atlases have played a pivotal role in this field, evolving from traditional printed histology atlases to digital atlases incorporating diverse imaging datasets. Magnetic resonance imaging (MRI)-based brain atlases, also known as brain maps, have been employed in specific studies. However, the existence of numerous versions of MRI-based brain atlases has impeded their standardized application and widespread use, despite the consensus within the academic community regarding their significance in mice and rats. Furthermore, there is a dearth of comprehensive and systematic reviews on MRI-based brain atlases for rodents. This review aims to bridge this gap by providing a comprehensive overview of the advancements in MRI-based brain atlases for rodents, with a specific focus on mice and rats. It seeks to explore the advantages and disadvantages of histologically printed brain atlases in comparison to MRI brain atlases, delineate the standardized methods for creating MRI brain atlases, and summarize their primary applications in neuroscience research. Additionally, this review aims to assist researchers in selecting appropriate versions of MRI brain atlases for their studies or refining existing MRI brain atlas resources, thereby facilitating the development and widespread adoption of standardized MRI-based brain atlases in rodents.
Collapse
Affiliation(s)
- Xiaoyi Ma
- Department of Geriatrics, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yao Xing
- School of Information Science and Technology, Fudan University, Shanghai, 200433, China
- Wuhan United Imaging Life Science Instrument Co., Ltd., Wuhan, 430071, China
| | - Renkuan Zhai
- Wuhan United Imaging Life Science Instrument Co., Ltd., Wuhan, 430071, China
| | - Yingying Du
- Wuhan United Imaging Life Science Instrument Co., Ltd., Wuhan, 430071, China
| | - Huanhuan Yan
- Shenzhen United Imaging Research Institute of Innovative Medical Equipment, Shenzhen, 518048, China
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
5
|
Abstract
This paper provides an overview of the role of neuroinflammation in Alzheimer's disease and other neurodegenerative diseases, highlighting the potential of anti-inflammatory treatments to slow or prevent decline. This research focuses on the use of positron emission tomography (PET) imaging to visualize and quantify molecular brain changes in patients, specifically microglial activation and reactive astrogliosis. We discuss the development and application of several PET radioligands, including first-generation ligands like PK11195 and Ro5-4864, as well as second- and third-generation ligands such as [11C]PBR28, [18F]DPA-714, [18F]GE-180, and [11C]ER176. These ligands target the 18-kDa translocator protein (TSPO), which is overexpressed in activated microglia and upregulated in astrocytes. We also address the limitations of these ligands, such as low brain uptake, poor penetration of the blood-brain barrier, short half-life, and variable kinetic behavior. Furthermore, we demonstrate the impact of genetic polymorphisms on ligand binding.
Collapse
Affiliation(s)
- Maura Malpetti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal and Gothenburg, Sweden
| | - Matthias Brendel
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
| |
Collapse
|
6
|
Finze A, Biechele G, Rauchmann BS, Franzmeier N, Palleis C, Katzdobler S, Weidinger E, Guersel S, Schuster S, Harris S, Schmitt J, Beyer L, Gnörich J, Lindner S, Albert NL, Wetzel CH, Rupprecht R, Rominger A, Danek A, Burow L, Kurz C, Tato M, Utecht J, Papazov B, Zaganjori M, Trappmann LK, Goldhardt O, Grimmer T, Haeckert J, Janowitz D, Buerger K, Keeser D, Stoecklein S, Dietrich O, Morenas-Rodriguez E, Barthel H, Sabri O, Bartenstein P, Simons M, Haass C, Höglinger GU, Levin J, Perneczky R, Brendel M. Individual regional associations between Aβ-, tau- and neurodegeneration (ATN) with microglial activation in patients with primary and secondary tauopathies. Mol Psychiatry 2023; 28:4438-4450. [PMID: 37495886 PMCID: PMC10827660 DOI: 10.1038/s41380-023-02188-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 06/27/2023] [Accepted: 07/10/2023] [Indexed: 07/28/2023]
Abstract
β-amyloid (Aβ) and tau aggregation as well as neuronal injury and atrophy (ATN) are the major hallmarks of Alzheimer's disease (AD), and biomarkers for these hallmarks have been linked to neuroinflammation. However, the detailed regional associations of these biomarkers with microglial activation in individual patients remain to be elucidated. We investigated a cohort of 55 patients with AD and primary tauopathies and 10 healthy controls that underwent TSPO-, Aβ-, tau-, and perfusion-surrogate-PET, as well as structural MRI. Z-score deviations for 246 brain regions were calculated and biomarker contributions of Aβ (A), tau (T), perfusion (N1), and gray matter atrophy (N2) to microglial activation (TSPO, I) were calculated for each individual subject. Individual ATN-related microglial activation was correlated with clinical performance and CSF soluble TREM2 (sTREM2) concentrations. In typical and atypical AD, regional tau was stronger and more frequently associated with microglial activation when compared to regional Aβ (AD: βT = 0.412 ± 0.196 vs. βA = 0.142 ± 0.123, p < 0.001; AD-CBS: βT = 0.385 ± 0.176 vs. βA = 0.131 ± 0.186, p = 0.031). The strong association between regional tau and microglia reproduced well in primary tauopathies (βT = 0.418 ± 0.154). Stronger individual associations between tau and microglial activation were associated with poorer clinical performance. In patients with 4RT, sTREM2 levels showed a positive association with tau-related microglial activation. Tau pathology has strong regional associations with microglial activation in primary and secondary tauopathies. Tau and Aβ related microglial response indices may serve as a two-dimensional in vivo assessment of neuroinflammation in neurodegenerative diseases.
Collapse
Grants
- EXC 2145 SyNergy - ID 390857198 Deutsche Forschungsgemeinschaft (German Research Foundation)
- EXC 2155 - project number 39087428 Deutsche Forschungsgemeinschaft (German Research Foundation)
- HO2402/18-1 Deutsche Forschungsgemeinschaft (German Research Foundation)
- FOR-2858 project numbers 403161218, 421887978 and 422188432 Deutsche Forschungsgemeinschaft (German Research Foundation)
- 19063p Alzheimer Forschung Initiative (Alzheimer Forschung Initiative e.V.)
- GUH was additionally funded by the German Federal Ministry of Education and Research (BMBF, 01KU1403A EpiPD; 01EK1605A HitTau; 01DH18025 TauTherapy); European Joint Programme on Rare Diseases (Improve-PSP); VolkswagenStiftung (Niedersächsisches Vorab); Petermax-Müller Foundation (Etiology and Therapy of Synucleinopathies and Tauopathies). The Lüneburg Heritage and Friedrich-Baur-Stiftung have supported the work of CP. The Hirnliga e.V. supported recruitment and imaging of the ActiGliA cohort (Manfred-Strohscheer-Stiftung) by a grant to BSR and MB.
- TG received consulting fees from AbbVie, Alector, Anavex, Biogen, Eli Lilly, Functional Neuromodulation, Grifols, Iqvia, Noselab, Novo Nordisk, NuiCare, Orphanzyme, Roche Diagnostics, Roche Pharma, UCB, and Vivoryon; lecture fees from Grifols, Medical Tribune, Novo Nordisk, Roche Pharma, and Schwabe; and has received grants to his institution from Roche Diagnostics.
- CH collaborates with Denali Therapeutics. CH is chief advisor of ISAR Bioscience and a member of the advisory board of AviadoBio.
- Günter Höglinger participated in industry-sponsored research projects from Abbvie, Biogen, Biohaven, Novartis, Roche, Sanofi, UCB; serves as a consultant for Abbvie, Alzprotect, Aprineua, Asceneuron, Bial, Biogen, Biohaven, Kyowa Kirin, Lundbeck, Novartis, Retrotope, Roche, Sanofi, UCB; received honoraria for scientific presentations from Abbvie, Bayer Vital, Bial, Biogen, Bristol Myers Squibb, Kyowa Kirin, Roche, Teva, UCB, Zambon; holds a patent on Treatment of Synucleinopathies. United States Patent No.: US 10,918,628 B2: EP 17 787 904.6-1109 / 3 525 788; received publication royalties from Academic Press, Kohlhammer, and Thieme.
Collapse
Affiliation(s)
- Anika Finze
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Gloria Biechele
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Boris-Stephan Rauchmann
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
- NeuroImaging Core Unit Munich (NICUM), LMU University Hospital, LMU Munich, Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Carla Palleis
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sabrina Katzdobler
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Endy Weidinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Selim Guersel
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Schuster
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Stefanie Harris
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Julia Schmitt
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Johannes Gnörich
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Christian H Wetzel
- Department of Psychiatry and Psychotherapy, University Regensburg, Regensburg, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University Regensburg, Regensburg, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Nuclear Medicine, University Hospital, Inselspital Bern, Bern, Switzerland
| | - Adrian Danek
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lena Burow
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Carolin Kurz
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Maia Tato
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Julia Utecht
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Boris Papazov
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
- NeuroImaging Core Unit Munich (NICUM), LMU University Hospital, LMU Munich, Munich, Germany
| | - Mirlind Zaganjori
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lena-Katharina Trappmann
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Oliver Goldhardt
- Department of Psychiatry and Psychotherapy, Klinikum Rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Timo Grimmer
- Department of Psychiatry and Psychotherapy, Klinikum Rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Jan Haeckert
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, Augsburg, Germany
| | | | | | - Daniel Keeser
- NeuroImaging Core Unit Munich (NICUM), LMU University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sophia Stoecklein
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Olaf Dietrich
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | | | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mikael Simons
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Christian Haass
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Munich, Germany
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Johannes Levin
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Robert Perneczky
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK
- Sheffield Institute for Translational Neurosciences (SITraN), University of Sheffield, Sheffield, UK
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
| |
Collapse
|
7
|
Lana D, Magni G, Landucci E, Wenk GL, Pellegrini-Giampietro DE, Giovannini MG. Phenomic Microglia Diversity as a Druggable Target in the Hippocampus in Neurodegenerative Diseases. Int J Mol Sci 2023; 24:13668. [PMID: 37761971 PMCID: PMC10531074 DOI: 10.3390/ijms241813668] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/31/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Phenomics, the complexity of microglia phenotypes and their related functions compels the continuous study of microglia in disease animal models to find druggable targets for neurodegenerative disorders. Activation of microglia was long considered detrimental for neuron survival, but more recently it has become apparent that the real scenario of microglia morphofunctional diversity is far more complex. In this review, we discuss the recent literature on the alterations in microglia phenomics in the hippocampus of animal models of normal brain aging, acute neuroinflammation, ischemia, and neurodegenerative disorders, such as AD. Microglia undergo phenomic changes consisting of transcriptional, functional, and morphological changes that transform them into cells with different properties and functions. The classical subdivision of microglia into M1 and M2, two different, all-or-nothing states is too simplistic, and does not correspond to the variety of phenotypes recently discovered in the brain. We will discuss the phenomic modifications of microglia focusing not only on the differences in microglia reactivity in the diverse models of neurodegenerative disorders, but also among different areas of the brain. For instance, in contiguous and highly interconnected regions of the rat hippocampus, microglia show a differential, finely regulated, and region-specific reactivity, demonstrating that microglia responses are not uniform, but vary significantly from area to area in response to insults. It is of great interest to verify whether the differences in microglia reactivity may explain the differential susceptibility of different brain areas to insults, and particularly the higher sensitivity of CA1 pyramidal neurons to inflammatory stimuli. Understanding the spatiotemporal heterogeneity of microglia phenomics in health and disease is of paramount importance to find new druggable targets for the development of novel microglia-targeted therapies in different CNS disorders. This will allow interventions in three different ways: (i) by suppressing the pro-inflammatory properties of microglia to limit the deleterious effect of their activation; (ii) by modulating microglia phenotypic change to favor anti-inflammatory properties; (iii) by influencing microglia priming early in the disease process.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Giada Magni
- Institute of Applied Physics “Nello Carrara”, National Research Council (IFAC-CNR), Via Madonna del Piano 10, 50019 Florence, Italy;
| | - Elisa Landucci
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Gary L. Wenk
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA;
| | - Domenico Edoardo Pellegrini-Giampietro
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| |
Collapse
|
8
|
Palumbo G, Kunze LH, Oos R, Wind-Mark K, Lindner S, von Ungern-Sternberg B, Bartenstein P, Ziegler S, Brendel M. Longitudinal Studies on Alzheimer Disease Mouse Models with Multiple Tracer PET/CT: Application of Reduction and Refinement Principles in Daily Practice to Safeguard Animal Welfare during Progressive Aging. Animals (Basel) 2023; 13:1812. [PMID: 37531139 PMCID: PMC10251952 DOI: 10.3390/ani13111812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/26/2023] [Accepted: 05/27/2023] [Indexed: 08/03/2023] Open
Abstract
Longitudinal studies on mouse models related to Alzheimer disease (AD) pathology play an important role in the investigation of therapeutic targets to help pharmaceutical research in the development of new drugs and in the attempt of an early diagnosis that can contribute to improving people's quality of life. There are several advantages to enriching longitudinal studies in AD models with Positron Emission Tomography (PET); among these advantages, the possibility of following the principle of the 3Rs of animal welfare is fundamental. In this manuscript, good daily experimental practice focusing on animal welfare is described and commented upon, based on the experience attained from studies conducted in our Nuclear Medicine department.
Collapse
Affiliation(s)
- Giovanna Palumbo
- Department of Nuclear Medicine, University Hospital LMU Munich, Marchionini Strasse 15, 81377 Munich, Germany
| | - Lea Helena Kunze
- Department of Nuclear Medicine, University Hospital LMU Munich, Marchionini Strasse 15, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Rosel Oos
- Department of Nuclear Medicine, University Hospital LMU Munich, Marchionini Strasse 15, 81377 Munich, Germany
| | - Karin Wind-Mark
- Department of Nuclear Medicine, University Hospital LMU Munich, Marchionini Strasse 15, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital LMU Munich, Marchionini Strasse 15, 81377 Munich, Germany
| | | | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital LMU Munich, Marchionini Strasse 15, 81377 Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital LMU Munich, Marchionini Strasse 15, 81377 Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital LMU Munich, Marchionini Strasse 15, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| |
Collapse
|
9
|
Hemonnot-Girard AL, Meersseman C, Pastore M, Garcia V, Linck N, Rey C, Chebbi A, Jeanneteau F, Ginsberg SD, Lachuer J, Reynes C, Rassendren F, Hirbec H. Comparative analysis of transcriptome remodeling in plaque-associated and plaque-distant microglia during amyloid-β pathology progression in mice. J Neuroinflammation 2022; 19:234. [PMID: 36153535 PMCID: PMC9508749 DOI: 10.1186/s12974-022-02581-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 08/24/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Research in recent years firmly established that microglial cells play an important role in the pathogenesis of Alzheimer's disease (AD). In parallel, a series of studies showed that, under both homeostatic and pathological conditions, microglia are a heterogeneous cell population. In AD, amyloid-β (Aβ) plaque-associated microglia (PAM) display a clearly distinct phenotype compared to plaque-distant microglia (PCM), suggesting that these two microglia subtypes likely differently contribute to disease progression. So far, molecular characterization of PAM was performed indirectly using single cell RNA sequencing (scRNA-seq) approaches or based on markers that are supposedly up-regulated in this microglia subpopulation. METHODS In this study based on a well-characterized AD mouse model, we combined cell-specific laser capture microdissection and RNA-seq analysis to i) identify, without preconceived notions of the molecular and/or functional changes that would affect these cells, the genes and gene networks that are dysregulated in PAM or PCM at three critical stages of the disease, and ii) to investigate the potential contribution of both plaque-associated and plaque-distant microglia. RESULTS First, we established that our approach allows selective isolation of microglia, while preserving spatial information and preventing transcriptome changes induced by classical purification approaches. Then, we identified, in PAM and PCM subpopulations, networks of co-deregulated genes and analyzed their potential functional roles in AD. Finally, we investigated the dynamics of microglia transcriptomic remodeling at early, intermediate and late stages of the disease and validated select findings in postmortem human AD brain. CONCLUSIONS Our comprehensive study provides useful transcriptomic information regarding the respective contribution of PAM and PCM across the Aβ pathology progression. It highlights specific pathways that would require further study to decipher their roles across disease progression. It demonstrates that the proximity of microglia to Aβ-plaques dramatically alters the microglial transcriptome and reveals that these changes can have both positive and negative impacts on the surrounding cells. These opposing effects may be driven by local microglia heterogeneity also demonstrated by this study. Our approach leads to molecularly define the less well studied plaque-distant microglia. We show that plaque-distant microglia are not bystanders of the disease, although the transcriptomic changes are far less striking compared to what is observed in plaque-associated microglia. In particular, our results suggest they may be involved in Aβ oligomer detection and in Aβ-plaque initiation, with increased contribution as the disease progresses.
Collapse
Affiliation(s)
- Anne-Laure Hemonnot-Girard
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Lyon, France
| | - Cédric Meersseman
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Lyon, France
| | - Manuela Pastore
- Université de Montpellier, CNRS, INSERM, BioCampus UAR3426, Montpellier, France
| | - Valentin Garcia
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Lyon, France
| | - Nathalie Linck
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Lyon, France
| | - Catherine Rey
- ProfileXpert, SFR santé Lyon-Est, CNRS UMR-S3453, Inserm US7, Lyon, France
| | - Amine Chebbi
- ProfileXpert, SFR santé Lyon-Est, CNRS UMR-S3453, Inserm US7, Lyon, France
| | | | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New-York, USA
- Department of Psychiatry, Department of Neuroscience & Physiology, and the NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, USA
| | - Joël Lachuer
- University Lyon1, CRCL-Centre de Recherche en Cancérologie de Lyon-Inserm U1052-CNRS U5286, Lyon, France
- ProfileXpert, SFR santé Lyon-Est, CNRS UMR-S3453, Inserm US7, Lyon, France
| | - Christelle Reynes
- Université de Montpellier, CNRS, INSERM, BioCampus UAR3426, Montpellier, France
| | - François Rassendren
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Lyon, France
| | - Hélène Hirbec
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France.
- LabEx Ion Channel Science and Therapeutics, Lyon, France.
| |
Collapse
|
10
|
Rauchmann B, Brendel M, Franzmeier N, Trappmann L, Zaganjori M, Ersoezlue E, Morenas‐Rodriguez E, Guersel S, Burow L, Kurz C, Haeckert J, Tatò M, Utecht J, Papazov B, Pogarell O, Janowitz D, Buerger K, Ewers M, Palleis C, Weidinger E, Biechele G, Schuster S, Finze A, Eckenweber F, Rupprecht R, Rominger A, Goldhardt O, Grimmer T, Keeser D, Stoecklein S, Dietrich O, Bartenstein P, Levin J, Höglinger G, Perneczky R. Microglial activation and connectivity in Alzheimer's disease and aging. Ann Neurol 2022; 92:768-781. [DOI: 10.1002/ana.26465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Boris‐Stephan Rauchmann
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
- Department of Neuroradiology University Hospital LMU Munich Germany
| | - Matthias Brendel
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich Munich Germany
| | - Lena Trappmann
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Mirlind Zaganjori
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Ersin Ersoezlue
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Estrella Morenas‐Rodriguez
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich Munich Germany
| | - Selim Guersel
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
| | - Lena Burow
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Carolin Kurz
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Jan Haeckert
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics University of Augsburg, Bezirkskrankenhaus Augsburg Augsburg Germany
| | - Maia Tatò
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Julia Utecht
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Boris Papazov
- Department of Radiology University Hospital, LMU Munich Munich Germany
| | - Oliver Pogarell
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich Munich Germany
| | - Katharina Buerger
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich Munich Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
| | - Michael Ewers
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich Munich Germany
| | - Carla Palleis
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
- Department of Neurology University Hospital, LMU Munich Munich Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich Germany
| | - Endy Weidinger
- Department of Neurology University Hospital, LMU Munich Munich Germany
| | - Gloria Biechele
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
| | - Sebastian Schuster
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
| | - Anika Finze
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
| | - Florian Eckenweber
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy University of Regensburg Regensburg Germany
| | - Axel Rominger
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
- Department of Nuclear Medicine University of Bern, Inselspital Bern Switzerland
| | - Oliver Goldhardt
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar Technical University Munich Munich Germany
| | - Timo Grimmer
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar Technical University Munich Munich Germany
| | - Daniel Keeser
- Department of Radiology University Hospital, LMU Munich Munich Germany
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
- Department of Neuroradiology University Hospital LMU Munich Germany
| | - Sophia Stoecklein
- Department of Radiology University Hospital, LMU Munich Munich Germany
| | - Olaf Dietrich
- Department of Radiology University Hospital, LMU Munich Munich Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
- Department of Neurology University Hospital, LMU Munich Munich Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich Germany
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
- Department of Neurology Hannover Medical School Hannover Germany
| | - Robert Perneczky
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
- Ageing Epidemiology (AGE) Research Unit, School of Public Health Imperial College London London UK
- Munich Cluster for Systems Neurology (SyNergy), Munich Germany
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
| |
Collapse
|
11
|
Tok S, Maurin H, Delay C, Crauwels D, Manyakov NV, Van Der Elst W, Moechars D, Drinkenburg WHIM. Pathological and neurophysiological outcomes of seeding human-derived tau pathology in the APP-KI NL-G-F and NL-NL mouse models of Alzheimer's Disease. Acta Neuropathol Commun 2022; 10:92. [PMID: 35739575 PMCID: PMC9219251 DOI: 10.1186/s40478-022-01393-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/07/2022] [Indexed: 12/02/2022] Open
Abstract
The two main histopathological hallmarks that characterize Alzheimer’s Disease are the presence of amyloid plaques and neurofibrillary tangles. One of the current approaches to studying the consequences of amyloid pathology relies on the usage of transgenic animal models that incorporate the mutant humanized form of the amyloid precursor protein (hAPP), with animal models progressively developing amyloid pathology as they age. However, these mice models generally overexpress the hAPP protein to facilitate the development of amyloid pathology, which has been suggested to elicit pathological and neuropathological changes unrelated to amyloid pathology. In this current study, we characterized APP knock-in (APP-KI) animals, that do not overexpress hAPP but still develop amyloid pathology to understand the influence of protein overexpression. We also induced tau pathology via human-derived tau seeding material to understand the neurophysiological effects of amyloid and tau pathology. We report that tau-seeded APP-KI animals progressively develop tau pathology, exacerbated by the presence of amyloid pathology. Interestingly, older amyloid-bearing, tau-seeded animals exhibited more amyloid pathology in the entorhinal area, isocortex and hippocampus, but not thalamus, which appeared to correlate with impairments in gamma oscillations before seeding. Tau-seeded animals also featured immediate deficits in power spectra values and phase-amplitude indices in the hippocampus after seeding, with gamma power spectra deficits persisting in younger animals. Both deficits in hippocampal phase-amplitude coupling and gamma power differentiate tau-seeded, amyloid-positive animals from buffer controls. Based on our results, impairments in gamma oscillations appear to be strongly associated with the presence and development of amyloid and tau pathology, and may also be an indicator of neuropathology, network dysfunction, and even potential disposition to the future development of amyloid pathology.
Collapse
Affiliation(s)
- S Tok
- Department of Neuroscience, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium.,Groningen Institute for Evolutionary Life Sciences, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - H Maurin
- Department of Neuroscience, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - C Delay
- Department of Neuroscience, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - D Crauwels
- Department of Neuroscience, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - N V Manyakov
- Data Sciences, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - W Van Der Elst
- Quantitative Sciences Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - D Moechars
- Department of Neuroscience, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - W H I M Drinkenburg
- Department of Neuroscience, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium. .,Groningen Institute for Evolutionary Life Sciences, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
12
|
Young JW, Barback CV, Stolz LA, Groman SM, Vera DR, Hoh C, Kotta KK, Minassian A, Powell SB, Brody AL. MicroPET evidence for a hypersensitive neuroinflammatory profile of gp120 mouse model of HIV. Psychiatry Res Neuroimaging 2022; 321:111445. [PMID: 35101828 DOI: 10.1016/j.pscychresns.2022.111445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 02/07/2023]
Abstract
Despite increased survivability for people living with HIV (PLWH), HIV-related cognitive deficits persist. Determining biological mechanism(s) underlying abnormalities is critical to minimize the long-term impact of HIV. Positron emission tomography (PET) studies reveal that PLWH exhibit elevated neuroinflammation, potentially contributing to these problems. PLWH are hypersensitive to environmental insults that drive elevated inflammatory profiles. Gp120 is an envelope glycoprotein exposed on the surface of the HIV envelope which enables HIV entry into a cell contributing to HIV-related neurotoxicity. In vivo evidence for mice overexpressing gp120 (transgenic) mice exhibiting neuroinflammation remains unclear. Here, we conducted microPET imaging in gp120 transgenic and wildtype mice, using the radiotracer [(18)F]FEPPA (binds to the translocator protein expressed by activated microglial serving as a neuroinflammatory marker). Imaging was performed at baseline and 24 h after lipopolysaccharide (LPS; 5 mg/kg) treatment (endotoxin that triggers an immune response). Gp120 transgenic mice exhibited elevated [(18F)]FEPPA in response to LPS vs. wildtype mice throughout the brain including dorsal and ventral striata, hypothalamus, and hippocampus. Gp120 transgenic mice are hypersensitive to environmental inflammatory insults, consistent with PLWH, measurable in vivo. It remains to-be-determined whether this heightened sensitivity is connected to the behavioral abnormalities of these mice or sensitive to any treatments.
Collapse
Affiliation(s)
- Jared W Young
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, USA; Research Service, VA San Diego Healthcare System, San Diego, CA, USA.
| | - Christopher V Barback
- Department of Radiology, University of California, San Diego, La Jolla California; UCSD In Vivo Cancer and Molecular Imaging Program
| | - Louise A Stolz
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, USA
| | - Stephanie M Groman
- Department of Neuroscience, Medical Discovery Team on Addiction, University of Minnesota
| | - David R Vera
- Department of Radiology, University of California, San Diego, La Jolla California; UCSD In Vivo Cancer and Molecular Imaging Program
| | - Carl Hoh
- Department of Radiology, University of California, San Diego, La Jolla California; UCSD In Vivo Cancer and Molecular Imaging Program
| | - Kishore K Kotta
- Department of Radiology, University of California, San Diego, La Jolla California; UCSD In Vivo Cancer and Molecular Imaging Program
| | - Arpi Minassian
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, USA; Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Susan B Powell
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, USA; Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Arthur L Brody
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, USA; Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
13
|
Blume T, Deussing M, Biechele G, Peters F, Zott B, Schmidt C, Franzmeier N, Wind K, Eckenweber F, Sacher C, Shi Y, Ochs K, Kleinberger G, Xiang X, Focke C, Lindner S, Gildehaus FJ, Beyer L, von Ungern-Sternberg B, Bartenstein P, Baumann K, Adelsberger H, Rominger A, Cumming P, Willem M, Dorostkar MM, Herms J, Brendel M. Chronic PPARγ Stimulation Shifts Amyloidosis to Higher Fibrillarity but Improves Cognition. Front Aging Neurosci 2022; 14:854031. [PMID: 35431893 PMCID: PMC9007038 DOI: 10.3389/fnagi.2022.854031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/25/2022] [Indexed: 11/30/2022] Open
Abstract
We undertook longitudinal β-amyloid positron emission tomography (Aβ-PET) imaging as a translational tool for monitoring of chronic treatment with the peroxisome proliferator-activated receptor gamma (PPARγ) agonist pioglitazone in Aβ model mice. We thus tested the hypothesis this treatment would rescue from increases of the Aβ-PET signal while promoting spatial learning and preservation of synaptic density. Here, we investigated longitudinally for 5 months PS2APP mice (N = 23; baseline age: 8 months) and AppNL–G–F mice (N = 37; baseline age: 5 months) using Aβ-PET. Groups of mice were treated with pioglitazone or vehicle during the follow-up interval. We tested spatial memory performance and confirmed terminal PET findings by immunohistochemical and biochemistry analyses. Surprisingly, Aβ-PET and immunohistochemistry revealed a shift toward higher fibrillary composition of Aβ-plaques during upon chronic pioglitazone treatment. Nonetheless, synaptic density and spatial learning were improved in transgenic mice with pioglitazone treatment, in association with the increased plaque fibrillarity. These translational data suggest that a shift toward higher plaque fibrillarity protects cognitive function and brain integrity. Increases in the Aβ-PET signal upon immunomodulatory treatments targeting Aβ aggregation can thus be protective.
Collapse
Affiliation(s)
- Tanja Blume
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany
| | - Maximilian Deussing
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Gloria Biechele
- Department of Radiology, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Finn Peters
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany
| | - Benedikt Zott
- Institute of Neuroscience, Technical University of Munich, Munich, Germany
- Department of Diagnostic and Interventional Neuroradiology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Claudio Schmidt
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Karin Wind
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Florian Eckenweber
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Christian Sacher
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Yuan Shi
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany
| | - Katharina Ochs
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany
| | - Gernot Kleinberger
- Metabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), Ludwig Maximilian University of Munich, Munich, Germany
- ISAR Bioscience GmbH, Planegg, Germany
| | - Xianyuan Xiang
- Metabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), Ludwig Maximilian University of Munich, Munich, Germany
| | - Carola Focke
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Franz-Josef Gildehaus
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Barbara von Ungern-Sternberg
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Karlheinz Baumann
- Roche Pharma Research and Early Development, Neuroscience Discovery, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Helmuth Adelsberger
- Department of Radiology, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Axel Rominger
- SyNergy, Ludwig Maximilian University of Munich, Munich, Germany
- Department of Nuclear Medicine, Inselspital Bern, Bern, Switzerland
| | - Paul Cumming
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD, Australia
| | - Michael Willem
- Metabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), Ludwig Maximilian University of Munich, Munich, Germany
| | - Mario M. Dorostkar
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig Maximilian University of Munich, Munich, Germany
| | - Jochen Herms
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany
- SyNergy, Ludwig Maximilian University of Munich, Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig Maximilian University of Munich, Munich, Germany
| | - Matthias Brendel
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
- SyNergy, Ludwig Maximilian University of Munich, Munich, Germany
- *Correspondence: Matthias Brendel,
| |
Collapse
|
14
|
Ni R. Positron Emission Tomography in Animal Models of Alzheimer's Disease Amyloidosis: Translational Implications. Pharmaceuticals (Basel) 2021; 14:1179. [PMID: 34832961 PMCID: PMC8623863 DOI: 10.3390/ph14111179] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/13/2021] [Accepted: 11/15/2021] [Indexed: 12/30/2022] Open
Abstract
Animal models of Alzheimer's disease amyloidosis that recapitulate cerebral amyloid-beta pathology have been widely used in preclinical research and have greatly enabled the mechanistic understanding of Alzheimer's disease and the development of therapeutics. Comprehensive deep phenotyping of the pathophysiological and biochemical features in these animal models is essential. Recent advances in positron emission tomography have allowed the non-invasive visualization of the alterations in the brain of animal models and in patients with Alzheimer's disease. These tools have facilitated our understanding of disease mechanisms and provided longitudinal monitoring of treatment effects in animal models of Alzheimer's disease amyloidosis. In this review, we focus on recent positron emission tomography studies of cerebral amyloid-beta accumulation, hypoglucose metabolism, synaptic and neurotransmitter receptor deficits (cholinergic and glutamatergic system), blood-brain barrier impairment, and neuroinflammation (microgliosis and astrocytosis) in animal models of Alzheimer's disease amyloidosis. We further propose the emerging targets and tracers for reflecting the pathophysiological changes and discuss outstanding challenges in disease animal models and future outlook in the on-chip characterization of imaging biomarkers towards clinical translation.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, ETH & University of Zurich, 8093 Zurich, Switzerland;
- Institute for Regenerative Medicine, University of Zurich, 8952 Zurich, Switzerland
| |
Collapse
|
15
|
Zhou R, Ji B, Kong Y, Qin L, Ren W, Guan Y, Ni R. PET Imaging of Neuroinflammation in Alzheimer's Disease. Front Immunol 2021; 12:739130. [PMID: 34603323 PMCID: PMC8481830 DOI: 10.3389/fimmu.2021.739130] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 08/27/2021] [Indexed: 12/15/2022] Open
Abstract
Neuroinflammation play an important role in Alzheimer's disease pathogenesis. Advances in molecular imaging using positron emission tomography have provided insights into the time course of neuroinflammation and its relation with Alzheimer's disease central pathologies in patients and in animal disease models. Recent single-cell sequencing and transcriptomics indicate dynamic disease-associated microglia and astrocyte profiles in Alzheimer's disease. Mitochondrial 18-kDa translocator protein is the most widely investigated target for neuroinflammation imaging. New generation of translocator protein tracers with improved performance have been developed and evaluated along with tau and amyloid imaging for assessing the disease progression in Alzheimer's disease continuum. Given that translocator protein is not exclusively expressed in glia, alternative targets are under rapid development, such as monoamine oxidase B, matrix metalloproteinases, colony-stimulating factor 1 receptor, imidazoline-2 binding sites, cyclooxygenase, cannabinoid-2 receptor, purinergic P2X7 receptor, P2Y12 receptor, the fractalkine receptor, triggering receptor expressed on myeloid cells 2, and receptor for advanced glycation end products. Promising targets should demonstrate a higher specificity for cellular locations with exclusive expression in microglia or astrocyte and activation status (pro- or anti-inflammatory) with highly specific ligand to enable in vivo brain imaging. In this review, we summarised recent advances in the development of neuroinflammation imaging tracers and provided an outlook for promising targets in the future.
Collapse
Affiliation(s)
- Rong Zhou
- Department of Nephrology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bin Ji
- Department of Radiopharmacy and Molecular Imaging, School of Pharmacy, Fudan University, Shanghai, China
| | - Yanyan Kong
- Positron Emission Tomography (PET) Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Limei Qin
- Inner Mongolia Baicaotang Qin Chinese Mongolia Hospital, Hohhot, China
| | - Wuwei Ren
- School of Information Science and Technology, Shanghaitech University, Shanghai, China
| | - Yihui Guan
- Positron Emission Tomography (PET) Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, University of Zurich & Eidgenössische Technische Hochschule Zürich (ETH Zurich), Zurich, Switzerland
| |
Collapse
|
16
|
Tournier BB, Tsartsalis S, Ceyzériat K, Fraser BH, Grégoire MC, Kövari E, Millet P. Astrocytic TSPO Upregulation Appears Before Microglial TSPO in Alzheimer's Disease. J Alzheimers Dis 2021; 77:1043-1056. [PMID: 32804124 PMCID: PMC7683091 DOI: 10.3233/jad-200136] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background: In vivo PET/SPECT imaging of neuroinflammation is primarily based on the estimation of the 18 kDa-translocator-protein (TSPO). However, TSPO is expressed by different cell types which complicates the interpretation. Objective: The present study evaluates the cellular origin of TSPO alterations in Alzheimer’s disease (AD). Methods: The TSPO cell origin was evaluated by combining radioactive imaging approaches using the TSPO radiotracer [125I]CLINDE and fluorescence-activated cell sorting, in a rat model of AD (TgF344-AD) and in AD subjects. Results: In the hippocampus of TgF344-AD rats, TSPO overexpression not only concerns glial cells but the increase is visible at 12 and 24 months in astrocytes and only at 24 months in microglia. In the temporal cortex of AD subjects, TSPO upregulation involved only glial cells. However, the mechanism of this upregulation appears different with an increase in the number of TSPO binding sites per cell without cell proliferation in the rat, and a microglial cell population expansion with a constant number of binding sites per cell in human AD. Conclusion: These data indicate an earlier astrocyte intervention than microglia and that TSPO in AD probably is an exclusive marker of glial activity without interference from other TSPO-expressing cells. This observation indicates that the interpretation of TSPO imaging depends on the stage of the pathology, and highlights the particular role of astrocytes.
Collapse
Affiliation(s)
- Benjamin B Tournier
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Switzerland
| | - Stergios Tsartsalis
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Switzerland
| | - Kelly Ceyzériat
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Switzerland.,Division of Nuclear medicine, University Hospitals of Geneva, Switzerland
| | - Ben H Fraser
- ANSTO LifeSciences, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Sydney, NSW, Australia
| | - Marie-Claude Grégoire
- ANSTO LifeSciences, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Sydney, NSW, Australia
| | - Enikö Kövari
- Division of Geriatric Psychiatry, Department of Mental Health and Psychiatry, University Hospitals of Geneva, Switzerland.,Department of Psychiatry, University of Geneva, Switzerland
| | - Philippe Millet
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Switzerland.,Department of Psychiatry, University of Geneva, Switzerland
| |
Collapse
|
17
|
Xu A, Tang Y, Zeng Q, Wang X, Tian H, Zhou Y, Li Z. Electroacupuncture Enhances Cognition by Promoting Brain Glucose Metabolism and Inhibiting Inflammation in the APP/PS1 Mouse Model of Alzheimer's Disease: A Pilot Study. J Alzheimers Dis 2021; 77:387-400. [PMID: 32741819 DOI: 10.3233/jad-200242] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disease, yet there is no effective treatment. Electroacupuncture (EA) is a complementary alternative medicine approach. In clinical and animal studies, EA promotes cognition in AD and vascular dementia. It has been previously reported that cognitive decline in AD might be closely related to reduced glucose intake in the brain. It is worth mentioning that the regions of glucose hypometabolism are usually found to be associated with neuroinflammation. OBJECTIVE This study is to explore whether the protective mechanism of EA on cognition is related to the regulation of glucose metabolism and neuroinflammation. METHODS APP/PS1 mice were randomly divided into AD group and the treatment (AD + EA) group. In the AD + EA group, EA was applied on Baihui (GV20) and Yintang (GV29) for 20 min and then pricked at Shuigou (GV26), once every alternate day for 4 weeks. Morris water maze (MWM) tests were performed to evaluate the effects of EA treatment on cognitive functions. 18F-FDG PET, immunofluorescence, and western blot were used to examine the mechanisms underlying EA effects. RESULTS From MWM tests, EA treatment significantly improved cognition of APP/PS1 mice. From the 18F-FDG PET, the levels of uptake rate of glucose in frontal lobe were higher than the AD group after EA. From immunofluorescence and western blot, amyloid-β (Aβ) and neuroinflammation were reduced after EA. CONCLUSION These results suggest that EA may prevent cognitive decline in AD mouse models by enhancing glucose metabolism and inhibiting inflammation-mediated Aβ deposition in the frontal lobe.
Collapse
Affiliation(s)
- Anping Xu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yinshan Tang
- Department of Rehabilitation and Traditional Chinese Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qingtao Zeng
- Information Engineering Institute, Beijing Institute of Graphic Communication, Beijing, China
| | - Xin Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Huiling Tian
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - You Zhou
- Department of Rehabilitation and Traditional Chinese Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhigang Li
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
18
|
Biechele G, Blume T, Deussing M, Zott B, Shi Y, Xiang X, Franzmeier N, Kleinberger G, Peters F, Ochs K, Focke C, Sacher C, Wind K, Schmidt C, Lindner S, Gildehaus FJ, Eckenweber F, Beyer L, von Ungern-Sternberg B, Bartenstein P, Baumann K, Dorostkar MM, Rominger A, Cumming P, Willem M, Adelsberger H, Herms J, Brendel M. Pre-therapeutic microglia activation and sex determine therapy effects of chronic immunomodulation. Theranostics 2021; 11:8964-8976. [PMID: 34522221 PMCID: PMC8419052 DOI: 10.7150/thno.64022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/06/2021] [Indexed: 12/21/2022] Open
Abstract
Modulation of the innate immune system is emerging as a promising therapeutic strategy against Alzheimer's disease (AD). However, determinants of a beneficial therapeutic effect are ill-understood. Thus, we investigated the potential of 18 kDa translocator protein positron-emission-tomography (TSPO-PET) for assessment of microglial activation in mouse brain before and during chronic immunomodulation. Methods: Serial TSPO-PET was performed during five months of chronic microglia modulation by stimulation of the peroxisome proliferator-activated receptor (PPAR)-γ with pioglitazone in two different mouse models of AD (PS2APP, AppNL-G-F ). Using mixed statistical models on longitudinal TSPO-PET data, we tested for effects of therapy and sex on treatment response. We tested correlations of baseline with longitudinal measures of TSPO-PET, and correlations between PET results with spatial learning performance and β-amyloid accumulation of individual mice. Immunohistochemistry was used to determine the molecular source of the TSPO-PET signal. Results: Pioglitazone-treated female PS2APP and AppNL-G-F mice showed attenuation of the longitudinal increases in TSPO-PET signal when compared to vehicle controls, whereas treated male AppNL-G-F mice showed the opposite effect. Baseline TSPO-PET strongly predicted changes in microglial activation in treated mice (R = -0.874, p < 0.0001) but not in vehicle controls (R = -0.356, p = 0.081). Reduced TSPO-PET signal upon pharmacological treatment was associated with better spatial learning despite higher fibrillar β-amyloid accumulation. Immunohistochemistry confirmed activated microglia to be the source of the TSPO-PET signal (R = 0.952, p < 0.0001). Conclusion: TSPO-PET represents a sensitive biomarker for monitoring of immunomodulation and closely reflects activated microglia. Sex and pre-therapeutic assessment of baseline microglial activation predict individual immunomodulation effects and may serve for responder stratification.
Collapse
Affiliation(s)
- Gloria Biechele
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Tanja Blume
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| | - Maximilian Deussing
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Benedikt Zott
- Institute of Neuroscience, Technical University of Munich, Munich, Germany
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Yuan Shi
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| | - Xianyuan Xiang
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | | | - Finn Peters
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| | - Katharina Ochs
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| | - Carola Focke
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Christian Sacher
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Karin Wind
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Claudio Schmidt
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Simon Lindner
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Franz-Josef Gildehaus
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Florian Eckenweber
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Leonie Beyer
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | | | - Peter Bartenstein
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- SyNergy, University of Munich, Munich, Germany
| | - Karlheinz Baumann
- Roche Pharma Research and Early Development, Neuroscience Discovery, Roche, Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Mario M. Dorostkar
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Axel Rominger
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- SyNergy, University of Munich, Munich, Germany
- Dept. of Nuclear Medicine, Inselspital Bern, Bern, Switzerland
| | - Paul Cumming
- Dept. of Nuclear Medicine, Inselspital Bern, Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, Australia
| | - Michael Willem
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Jochen Herms
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
- SyNergy, University of Munich, Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Matthias Brendel
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- SyNergy, University of Munich, Munich, Germany
| |
Collapse
|
19
|
Zhang PF, Hu H, Tan L, Yu JT. Microglia Biomarkers in Alzheimer's Disease. Mol Neurobiol 2021; 58:3388-3404. [PMID: 33713018 DOI: 10.1007/s12035-021-02348-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 03/03/2021] [Indexed: 12/13/2022]
Abstract
Early detection and clinical diagnosis of Alzheimer's disease (AD) have become an extremely important link in the prevention and treatment of AD. Because of the occult onset, the diagnosis and treatment of AD based on clinical symptoms are increasingly challenged by current severe situations. Therefore, molecular diagnosis models based on early AD pathological markers have received more attention. Among the possible pathological mechanisms, microglia which are necessary for normal brain function are highly expected and have been continuously studied in various models. Several AD biomarkers already exist, but currently there is a paucity of specific and sensitive microglia biomarkers which can accurately measure preclinical AD. Bringing microglia biomarkers into the molecular diagnostic system which is based on fluid and neuroimaging will play an important role in future scientific research and clinical practice. Furthermore, developing novel, more specific, and sensitive microglia biomarkers will make it possible to pharmaceutically target chemical pathways that preserve beneficial microglial functions in response to AD pathology. This review discusses microglia biomarkers in the context of AD.
Collapse
Affiliation(s)
- Peng-Fei Zhang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
20
|
Jung ME. A Protective Role of Translocator Protein in Alzheimer's Disease Brain. Curr Alzheimer Res 2021; 17:3-15. [PMID: 32065102 DOI: 10.2174/1567205017666200217105950] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 01/13/2020] [Accepted: 01/20/2020] [Indexed: 12/16/2022]
Abstract
Translocator Protein (18 kDa) (TSPO) is a mitochondrial protein that locates cytosol cholesterol to mitochondrial membranes to begin the synthesis of steroids including neurotrophic neurosteroids. TSPO is abundantly present in glial cells that support neurons and respond to neuroinflammation. Located at the outer membrane of mitochondria, TSPO regulates the opening of mitochondrial permeability transition pore (mPTP) that controls the entry of molecules necessary for mitochondrial function. TSPO is linked to neurodegenerative Alzheimer's Disease (AD) such that TSPO is upregulated in the brain of AD patients and signals AD-induced adverse changes in brain. The initial increase in TSPO in response to brain insults remains elevated to repair cellular damages and perhaps to prevent further neuronal degeneration as AD progresses. To exert such protective activities, TSPO increases the synthesis of neuroprotective steroids, decreases neuroinflammation, limits the opening of mPTP, and reduces the generation of reactive oxygen species. The beneficial effects of TSPO on AD brain are manifested as the attenuation of neurotoxic amyloid β and mitochondrial dysfunction accompanied by the improvement of memory and cognition. However, the protective activities of TSPO appear to be temporary and eventually diminish as the severity of AD becomes profound. Timely treatment with TSPO agonists/ligands before the loss of endogenous TSPO's activity may promote the protective functions and may extend neuronal survival.
Collapse
Affiliation(s)
- Marianna E Jung
- Pharmacology and Neuroscience, University of North Texas Health Science Center, Institute for Healthy Aging, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, United States
| |
Collapse
|
21
|
The Microbiota-Gut-Brain Axis and Alzheimer Disease. From Dysbiosis to Neurodegeneration: Focus on the Central Nervous System Glial Cells. J Clin Med 2021; 10:jcm10112358. [PMID: 34072107 PMCID: PMC8199461 DOI: 10.3390/jcm10112358] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 02/06/2023] Open
Abstract
The microbiota-gut system can be thought of as a single unit that interacts with the brain via the "two-way" microbiota-gut-brain axis. Through this axis, a constant interplay mediated by the several products originating from the microbiota guarantees the physiological development and shaping of the gut and the brain. In the present review will be described the modalities through which the microbiota and gut control each other, and the main microbiota products conditioning both local and brain homeostasis. Much evidence has accumulated over the past decade in favor of a significant association between dysbiosis, neuroinflammation and neurodegeneration. Presently, the pathogenetic mechanisms triggered by molecules produced by the altered microbiota, also responsible for the onset and evolution of Alzheimer disease, will be described. Our attention will be focused on the role of astrocytes and microglia. Numerous studies have progressively demonstrated how these glial cells are important to ensure an adequate environment for neuronal activity in healthy conditions. Furthermore, it is becoming evident how both cell types can mediate the onset of neuroinflammation and lead to neurodegeneration when subjected to pathological stimuli. Based on this information, the role of the major microbiota products in shifting the activation profiles of astrocytes and microglia from a healthy to a diseased state will be discussed, focusing on Alzheimer disease pathogenesis.
Collapse
|
22
|
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
|
23
|
Shin JH, Hwang YS, Jung BK, Seo SH, Ham DW, Shin EH. Reduction of Amyloid Burden by Proliferated Homeostatic Microglia in Toxoplasma gondii-Infected Alzheimer's Disease Model Mice. Int J Mol Sci 2021; 22:ijms22052764. [PMID: 33803262 PMCID: PMC7975980 DOI: 10.3390/ijms22052764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/26/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
In this study, we confirmed that the number of resident homeostatic microglia increases during chronic Toxoplasma gondii infection. Given that the progression of Alzheimer’s disease (AD) worsens with the accumulation of amyloid β (Aβ) plaques, which are eliminated through microglial phagocytosis, we hypothesized that T. gondii-induced microglial proliferation would reduce AD progression. Therefore, we investigated the association between microglial proliferation and Aβ plaque burden using brain tissues isolated from 5XFAD AD mice (AD group) and T. gondii-infected AD mice (AD + Toxo group). In the AD + Toxo group, amyloid plaque burden significantly decreased compared with the AD group; conversely, homeostatic microglial proliferation, and number of plaque-associated microglia significantly increased. As most plaque-associated microglia shifted to the disease-associated microglia (DAM) phenotype in both AD and AD + Toxo groups and underwent apoptosis after the lysosomal degradation of phagocytosed Aβ plaques, this indicates that a sustained supply of homeostatic microglia is required for alleviating Aβ plaque burden. Thus, chronic T. gondii infection can induce microglial proliferation in the brains of mice with progressed AD; a sustained supply of homeostatic microglia is a promising prospect for AD treatment.
Collapse
Affiliation(s)
- Ji-Hun Shin
- Department of Tropical Medicine and Parasitology, Seoul National University College of Medicine, and Institute of Endemic Diseases, Seoul 03080, Korea; (J.-H.S.); (Y.S.H.); (S.-H.S.); (D.-W.H.)
| | - Young Sang Hwang
- Department of Tropical Medicine and Parasitology, Seoul National University College of Medicine, and Institute of Endemic Diseases, Seoul 03080, Korea; (J.-H.S.); (Y.S.H.); (S.-H.S.); (D.-W.H.)
| | - Bong-Kwang Jung
- Institute of Parasitic Diseases, Korea Association of Health Promotion, Seoul 07649, Korea;
| | - Seung-Hwan Seo
- Department of Tropical Medicine and Parasitology, Seoul National University College of Medicine, and Institute of Endemic Diseases, Seoul 03080, Korea; (J.-H.S.); (Y.S.H.); (S.-H.S.); (D.-W.H.)
| | - Do-Won Ham
- Department of Tropical Medicine and Parasitology, Seoul National University College of Medicine, and Institute of Endemic Diseases, Seoul 03080, Korea; (J.-H.S.); (Y.S.H.); (S.-H.S.); (D.-W.H.)
| | - Eun-Hee Shin
- Department of Tropical Medicine and Parasitology, Seoul National University College of Medicine, and Institute of Endemic Diseases, Seoul 03080, Korea; (J.-H.S.); (Y.S.H.); (S.-H.S.); (D.-W.H.)
- Seoul National University Bundang Hospital, Seongnam 13620, Korea
- Correspondence: ; Tel.: +82-2-740-8344
| |
Collapse
|
24
|
Behrangi N, Lorenz P, Kipp M. Oligodendrocyte Lineage Marker Expression in eGFP-GFAP Transgenic Mice. J Mol Neurosci 2020; 71:2237-2248. [PMID: 33346907 PMCID: PMC8585802 DOI: 10.1007/s12031-020-01771-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/02/2020] [Indexed: 12/21/2022]
Abstract
Oligodendrocytes, the myelinating cells of the central nervous system, orchestrate several key cellular functions in the brain and spinal cord, including axon insulation, energy transfer to neurons, and, eventually, modulation of immune responses. There is growing interest for obtaining reliable markers that can specifically label oligodendroglia and their progeny. In many studies, anti-CC1 antibodies, presumably recognizing the protein adenomatous polyposis coli (APC), are used to label mature, myelinating oligodendrocytes. However, it has been discussed whether anti-CC1 antibodies could recognize as well, under pathological conditions, other cell populations, particularly astrocytes. In this study, we used transgenic mice in which astrocytes are labeled by the enhanced green fluorescent protein (eGFP) under the control of the human glial fibrillary acidic protein (GFAP) promoter. By detailed co-localization studies we were able to demonstrate that a significant proportion of eGFP-expressing cells co-express markers of the oligodendrocyte lineage, such as the transcription factor Oligodendrocyte Transcription Factor 2 (OLIG2); the NG2 proteoglycan, also known as chrondroitin sulfate proteoglycan 4 (CSPG4); or APC. The current finding that the GFAP promoter drives transgene expression in cells of the oligodendrocyte lineage should be considered when interpreting results from co-localization studies.
Collapse
Affiliation(s)
- Newshan Behrangi
- Institute of Anatomy, Rostock University Medical Center, 18057, Rostock, Germany.,Department of Anatomy II, Ludwig-Maximilians-University of Munich, 80336, Munich, Germany
| | - Peter Lorenz
- Institute of Immunology, Rostock University Medical Center, 18057, Rostock, Germany
| | - Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, 18057, Rostock, Germany. .,Center for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Center, Gelsheimer Strasse 20, 18147, Rostock, Germany.
| |
Collapse
|
25
|
Biechele G, Franzmeier N, Blume T, Ewers M, Luque JM, Eckenweber F, Sacher C, Beyer L, Ruch-Rubinstein F, Lindner S, Gildehaus FJ, von Ungern-Sternberg B, Cumming P, Bartenstein P, Rominger A, Höglinger GU, Herms J, Brendel M. Glial activation is moderated by sex in response to amyloidosis but not to tau pathology in mouse models of neurodegenerative diseases. J Neuroinflammation 2020; 17:374. [PMID: 33317543 PMCID: PMC7737385 DOI: 10.1186/s12974-020-02046-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022] Open
Abstract
Background In vivo assessment of neuroinflammation by 18-kDa translocator protein positron-emission-tomography (TSPO-PET) ligands receives growing interest in preclinical and clinical research of neurodegenerative disorders. Higher TSPO-PET binding as a surrogate for microglial activation in females has been reported for cognitively normal humans, but such effects have not yet been evaluated in rodent models of neurodegeneration and their controls. Thus, we aimed to investigate the impact of sex on microglial activation in amyloid and tau mouse models and wild-type controls. Methods TSPO-PET (18F-GE-180) data of C57Bl/6 (wild-type), AppNL-G-F (β-amyloid model), and P301S (tau model) mice was assessed longitudinally between 2 and 12 months of age. The AppNL-G-F group also underwent longitudinal β-amyloid-PET imaging (Aβ-PET; 18F-florbetaben). PET results were confirmed and validated by immunohistochemical investigation of microglial (Iba-1, CD68), astrocytic (GFAP), and tau (AT8) markers. Findings in cerebral cortex were compared by sex using linear mixed models for PET data and analysis of variance for immunohistochemistry. Results Wild-type mice showed an increased TSPO-PET signal over time (female +23%, male +4%), with a significant sex × age interaction (T = − 4.171, p < 0.001). The Aβ model AppNL-G-F mice also showed a significant sex × age interaction (T = − 2.953, p = 0.0048), where cortical TSPO-PET values increased by 31% in female AppNL-G-F mice, versus only 6% in the male mice group from 2.5 to 10 months of age. Immunohistochemistry for the microglial markers Iba-1 and CD68 confirmed the TSPO-PET findings in male and female mice aged 10 months. Aβ-PET in the same AppNL-G-F mice indicated no significant sex × age interaction (T = 0.425, p = 0.673). The P301S tau model showed strong cortical increases of TSPO-PET from 2 to 8.5 months of age (female + 32%, male + 36%), without any significant sex × age interaction (T = − 0.671, p = 0.504), and no sex differences in Iba-1, CD68, or AT8 immunohistochemistry. Conclusion Female mice indicate sex-dependent microglia activation in aging and in response to amyloidosis but not in response to tau pathology. This calls for consideration of sex difference in TSPO-PET studies of microglial activation in mouse models of neurodegeneration and by extension in human studies.
Collapse
Affiliation(s)
- Gloria Biechele
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Tanja Blume
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany.,DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| | - Michael Ewers
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.,DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| | - Jose Medina Luque
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| | - Florian Eckenweber
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Christian Sacher
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Francois Ruch-Rubinstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Franz-Josef Gildehaus
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Barbara von Ungern-Sternberg
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Paul Cumming
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland.,School of Psychology and Counselling, Queensland University of Technology, Brisbane, Australia
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany.,Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany.,Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Günter U Höglinger
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.,Department of Neurology, Technical University Munich, Munich, Germany.,Center of Neuropathology and Prion Research, University of Munich, Munich, Germany
| | - Jochen Herms
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany. .,Department of Neurology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
26
|
Lana D, Ugolini F, Giovannini MG. Space-Dependent Glia-Neuron Interplay in the Hippocampus of Transgenic Models of β-Amyloid Deposition. Int J Mol Sci 2020; 21:E9441. [PMID: 33322419 PMCID: PMC7763751 DOI: 10.3390/ijms21249441] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 12/14/2022] Open
Abstract
This review is focused on the description and discussion of the alterations of astrocytes and microglia interplay in models of Alzheimer's disease (AD). AD is an age-related neurodegenerative pathology with a slowly progressive and irreversible decline of cognitive functions. One of AD's histopathological hallmarks is the deposition of amyloid beta (Aβ) plaques in the brain. Long regarded as a non-specific, mere consequence of AD pathology, activation of microglia and astrocytes is now considered a key factor in both initiation and progression of the disease, and suppression of astrogliosis exacerbates neuropathology. Reactive astrocytes and microglia overexpress many cytokines, chemokines, and signaling molecules that activate or damage neighboring cells and their mutual interplay can result in virtuous/vicious cycles which differ in different brain regions. Heterogeneity of glia, either between or within a particular brain region, is likely to be relevant in healthy conditions and disease processes. Differential crosstalk between astrocytes and microglia in CA1 and CA3 areas of the hippocampus can be responsible for the differential sensitivity of the two areas to insults. Understanding the spatial differences and roles of glia will allow us to assess how these interactions can influence the state and progression of the disease, and will be critical for identifying therapeutic strategies.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy;
| | - Filippo Ugolini
- Department of Health Sciences, Section of Anatomopathology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy;
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy;
| |
Collapse
|
27
|
Lisova K, Wang J, Chao PH, van Dam RM. A simple and efficient automated microvolume radiosynthesis of [ 18F]Florbetaben. EJNMMI Radiopharm Chem 2020; 5:30. [PMID: 33275179 PMCID: PMC7718361 DOI: 10.1186/s41181-020-00113-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Current automated radiosynthesizers are generally optimized for producing large batches of PET tracers. Preclinical imaging studies, however, often require only a small portion of a regular batch, which cannot be economically produced on a conventional synthesizer. Alternative approaches are desired to produce small to moderate batches to reduce cost and the amount of reagents and radioisotope needed to produce PET tracers with high molar activity. In this work we describe the first reported microvolume method for production of [18F]Florbetaben for use in imaging of Alzheimer's disease. PROCEDURES The microscale synthesis of [18F]Florbetaben was adapted from conventional-scale synthesis methods. Aqueous [18F]fluoride was azeotropically dried with K2CO3/K222 (275/383 nmol) complex prior to radiofluorination of the Boc-protected precursor (80 nmol) in 10 μL DMSO at 130 °C for 5 min. The resulting intermediate was deprotected with HCl at 90 °C for 3 min and recovered from the chip in aqueous acetonitrile solution. The crude product was purified via analytical scale HPLC and the collected fraction reformulated via solid-phase extraction using a miniature C18 cartridge. RESULTS Starting with 270 ± 100 MBq (n = 3) of [18F]Fluoride, the method affords formulated product with 49 ± 3% (decay-corrected) yield,> 98% radiochemical purity and a molar activity of 338 ± 55 GBq/μmol. The miniature C18 cartridge enables efficient elution with only 150 μL of ethanol which is diluted to a final volume of 1.0 mL, thus providing a sufficient concentration for in vivo imaging. The whole procedure can be completed in 55 min. CONCLUSIONS This work describes an efficient and reliable procedure to produce [18F]Florbetaben in quantities sufficient for large-scale preclinical applications. This method provides very high yields and molar activities compared to reported literature methods. This method can be applied to higher starting activities with special consideration given to automation and radiolysis prevention.
Collapse
Affiliation(s)
- Ksenia Lisova
- Physics & Biology in Medicine Interdepartmental Graduate Program, University of California Los Angeles, Los Angeles, CA, USA
- Crump Institute for Molecular Imaging, University of California Los Angeles, Los Angeles, CA, USA
- Department of Molecular & Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Jia Wang
- Crump Institute for Molecular Imaging, University of California Los Angeles, Los Angeles, CA, USA
- Department of Molecular & Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Philip H Chao
- Crump Institute for Molecular Imaging, University of California Los Angeles, Los Angeles, CA, USA
- Department of Molecular & Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - R Michael van Dam
- Physics & Biology in Medicine Interdepartmental Graduate Program, University of California Los Angeles, Los Angeles, CA, USA.
- Crump Institute for Molecular Imaging, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Molecular & Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
28
|
Palleis C, Sauerbeck J, Beyer L, Harris S, Schmitt J, Morenas-Rodriguez E, Finze A, Nitschmann A, Ruch-Rubinstein F, Eckenweber F, Biechele G, Blume T, Shi Y, Weidinger E, Prix C, Bötzel K, Danek A, Rauchmann BS, Stöcklein S, Lindner S, Unterrainer M, Albert NL, Wetzel C, Rupprecht R, Rominger A, Bartenstein P, Herms J, Perneczky R, Haass C, Levin J, Höglinger GU, Brendel M. In Vivo Assessment of Neuroinflammation in 4-Repeat Tauopathies. Mov Disord 2020; 36:883-894. [PMID: 33245166 DOI: 10.1002/mds.28395] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/28/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Neuroinflammation has received growing interest as a therapeutic target in neurodegenerative disorders, including 4-repeat tauopathies. OBJECTIVES The aim of this cross-sectional study was to investigate 18 kDa translocator protein positron emission tomography (PET) as a biomarker for microglial activation in the 4-repeat tauopathies corticobasal degeneration and progressive supranuclear palsy. METHODS Specific binding of the 18 kDa translocator protein tracer 18 F-GE-180 was determined by serial PET during pharmacological depletion of microglia in a 4-repeat tau mouse model. The 18 kDa translocator protein PET was performed in 30 patients with corticobasal syndrome (68 ± 9 years, 16 women) and 14 patients with progressive supranuclear palsy (69 ± 9 years, 8 women), and 13 control subjects (70 ± 7 years, 7 women). Group comparisons and associations with parameters of disease progression were assessed by region-based and voxel-wise analyses. RESULTS Tracer binding was significantly reduced after pharmacological depletion of microglia in 4-repeat tau mice. Elevated 18 kDa translocator protein labeling was observed in the subcortical brain areas of patients with corticobasal syndrome and progressive supranuclear palsy when compared with controls and was most pronounced in the globus pallidus internus, whereas only patients with corticobasal syndrome showed additionally elevated tracer binding in motor and supplemental motor areas. The 18 kDa translocator protein labeling was not correlated with parameters of disease progression in corticobasal syndrome and progressive supranuclear palsy but allowed sensitive detection in patients with 4-repeat tauopathies by a multiregion classifier. CONCLUSIONS Our data indicate that 18 F-GE-180 PET detects microglial activation in the brain of patients with 4-repeat tauopathy, fitting to predilection sites of the phenotype. The 18 kDa translocator protein PET has a potential for monitoring neuroinflammation in 4-repeat tauopathies. © 2020 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Carla Palleis
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Julia Sauerbeck
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Stefanie Harris
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Julia Schmitt
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | | | - Anika Finze
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Alexander Nitschmann
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | | | - Florian Eckenweber
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Gloria Biechele
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Tanja Blume
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Yuan Shi
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Endy Weidinger
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Catharina Prix
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Kai Bötzel
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Adrian Danek
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Boris-Stephan Rauchmann
- Department of Radiology, University Hospital of Munich, LMU Munich, Munich, Germany.,Center for Neuropathology and Prion Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Sophia Stöcklein
- Department of Radiology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Marcus Unterrainer
- Department of Radiology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Christian Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.,Department of Nuclear Medicine, University of Bern, Inselspital, Bern, Switzerland
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.,Chair of Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases, Munich, Germany.,Center for Neuropathology and Prion Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases, Munich, Germany.,Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany.,Ageing Epidemiology Research Unit, School of Public Health, Imperial College, London, UK
| | - Christian Haass
- German Center for Neurodegenerative Diseases, Munich, Germany.,Department of Nuclear Medicine, University of Bern, Inselspital, Bern, Switzerland.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Johannes Levin
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany.,German Center for Neurodegenerative Diseases, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
29
|
Ewers M, Biechele G, Suárez-Calvet M, Sacher C, Blume T, Morenas-Rodriguez E, Deming Y, Piccio L, Cruchaga C, Kleinberger G, Shaw L, Trojanowski JQ, Herms J, Dichgans M, Brendel M, Haass C, Franzmeier N. Higher CSF sTREM2 and microglia activation are associated with slower rates of beta-amyloid accumulation. EMBO Mol Med 2020; 12:e12308. [PMID: 32790063 PMCID: PMC7507349 DOI: 10.15252/emmm.202012308] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 07/03/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
Microglia activation is the brain's major immune response to amyloid plaques in Alzheimer's disease (AD). Both cerebrospinal fluid (CSF) levels of soluble TREM2 (sTREM2), a biomarker of microglia activation, and microglia PET are increased in AD; however, whether an increase in these biomarkers is associated with reduced amyloid-beta (Aβ) accumulation remains unclear. To address this question, we pursued a two-pronged translational approach. Firstly, in non-demented and demented individuals, we tested CSF sTREM2 at baseline to predict (i) amyloid PET changes over ∼2 years and (ii) tau PET cross-sectionally assessed in a subset of patients. We found higher CSF sTREM2 associated with attenuated amyloid PET increase and lower tau PET. Secondly, in the AppNL-G-F mouse model of amyloidosis, we studied baseline 18 F-GE180 microglia PET and longitudinal amyloid PET to test the microglia vs. Aβ association, without any confounding co-pathologies often present in AD patients. Higher microglia PET at age 5 months was associated with a slower amyloid PET increase between ages 5-to-10 months. In conclusion, higher microglia activation as determined by CSF sTREM2 or microglia PET shows protective effects on subsequent amyloid accumulation.
Collapse
Affiliation(s)
- Michael Ewers
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University (LMU), Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Gloria Biechele
- Department of Nuclear Medicine, University Hospital Munich, Ludwig Maximilian University Munich, Munich, Germany
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Servei de Neurologia, Hospital del Mar, Barcelona, Spain
| | - Christian Sacher
- Department of Nuclear Medicine, University Hospital Munich, Ludwig Maximilian University Munich, Munich, Germany
| | - Tanja Blume
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Estrella Morenas-Rodriguez
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Yuetiva Deming
- Department of Population Health Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Laura Piccio
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.,Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
| | - Carlos Cruchaga
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.,Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Gernot Kleinberger
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany.,ISAR Bioscience GmbH, Planegg, Germany
| | - Leslie Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University (LMU), Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | | | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital Munich, Ludwig Maximilian University Munich, Munich, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University (LMU), Munich, Germany
| |
Collapse
|
30
|
Eckenweber F, Medina-Luque J, Blume T, Sacher C, Biechele G, Wind K, Deussing M, Briel N, Lindner S, Boening G, von Ungern-Sternberg B, Unterrainer M, Albert NL, Zwergal A, Levin J, Bartenstein P, Cumming P, Rominger A, Höglinger GU, Herms J, Brendel M. Longitudinal TSPO expression in tau transgenic P301S mice predicts increased tau accumulation and deteriorated spatial learning. J Neuroinflammation 2020; 17:208. [PMID: 32660586 PMCID: PMC7358201 DOI: 10.1186/s12974-020-01883-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/30/2020] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND P301S tau transgenic mice show age-dependent accumulation of neurofibrillary tangles in the brainstem, hippocampus, and neocortex, leading to neuronal loss and cognitive deterioration. However, there is hitherto only sparse documentation of the role of neuroinflammation in tau mouse models. Thus, we analyzed longitudinal microglial activation by small animal 18 kDa translocator protein positron-emission-tomography (TSPO μPET) imaging in vivo, in conjunction with terminal assessment of tau pathology, spatial learning, and cerebral glucose metabolism. METHODS Transgenic P301S (n = 33) and wild-type (n = 18) female mice were imaged by 18F-GE-180 TSPO μPET at the ages of 1.9, 3.9, and 6.4 months. We conducted behavioral testing in the Morris water maze, 18F-fluordesoxyglucose (18F-FDG) μPET, and AT8 tau immunohistochemistry at 6.3-6.7 months. Terminal microglial immunohistochemistry served for validation of TSPO μPET results in vivo, applying target regions in the brainstem, cortex, cerebellum, and hippocampus. We compared the results with our historical data in amyloid-β mouse models. RESULTS TSPO expression in all target regions of P301S mice increased exponentially from 1.9 to 6.4 months, leading to significant differences in the contrasts with wild-type mice at 6.4 months (+ 11-23%, all p < 0.001), but the apparent microgliosis proceeded more slowly than in our experience in amyloid-β mouse models. Spatial learning and glucose metabolism of AT8-positive P301S mice were significantly impaired at 6.3-6.5 months compared to the wild-type group. Longitudinal increases in TSPO expression predicted greater tau accumulation and lesser spatial learning performance at 6.3-6.7 months. CONCLUSIONS Monitoring of TSPO expression as a surrogate of microglial activation in P301S tau transgenic mice by μPET indicates a delayed time course when compared to amyloid-β mouse models. Detrimental associations of microglial activation with outcome parameters are opposite to earlier data in amyloid-β mouse models. The contribution of microglial response to pathology accompanying amyloid-β and tau over-expression merits further investigation.
Collapse
Affiliation(s)
- Florian Eckenweber
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
| | - Jose Medina-Luque
- Center of Neuropathology and Prion Research, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
| | - Tanja Blume
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
- Center of Neuropathology and Prion Research, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377, Munich, Germany
| | - Christian Sacher
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
| | - Gloria Biechele
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
| | - Karin Wind
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
| | - Maximilian Deussing
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
| | - Nils Briel
- Center of Neuropathology and Prion Research, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
| | - Guido Boening
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
| | | | - Marcus Unterrainer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
| | - Andreas Zwergal
- German Center for Vertigo and Balance Disorders, DSGZ, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
- Department of Neurology, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377, Munich, Germany
- Department of Neurology, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany
| | - Paul Cumming
- Department of Nuclear Medicine, Inselspital Bern, Bern, Switzerland
- School of Psychology and Counselling and IHBI, Queensland University of Technology, Brisbane, Australia
| | - Axel Rominger
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany
- Department of Nuclear Medicine, Inselspital Bern, Bern, Switzerland
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377, Munich, Germany
- Department of Neurology, Hannover Medical School, Hannover, Germany
- Department of Neurology, Technical University of Munich, Munich, Germany
| | - Jochen Herms
- Center of Neuropathology and Prion Research, University Hospital of Munich, LMU Munich, 81377, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany.
| |
Collapse
|
31
|
Sacher C, Blume T, Beyer L, Biechele G, Sauerbeck J, Eckenweber F, Deussing M, Focke C, Parhizkar S, Lindner S, Gildehaus FJ, von Ungern-Sternberg B, Baumann K, Tahirovic S, Kleinberger G, Willem M, Haass C, Bartenstein P, Cumming P, Rominger A, Herms J, Brendel M. Asymmetry of Fibrillar Plaque Burden in Amyloid Mouse Models. J Nucl Med 2020; 61:1825-1831. [DOI: 10.2967/jnumed.120.242750] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/03/2020] [Indexed: 11/16/2022] Open
|
32
|
Rao CV, Asch AS, Carr DJJ, Yamada HY. "Amyloid-beta accumulation cycle" as a prevention and/or therapy target for Alzheimer's disease. Aging Cell 2020; 19:e13109. [PMID: 31981470 PMCID: PMC7059149 DOI: 10.1111/acel.13109] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/16/2019] [Accepted: 12/25/2019] [Indexed: 02/06/2023] Open
Abstract
The cell cycle and its regulators are validated targets for cancer drugs. Reagents that target cells in a specific cell cycle phase (e.g., antimitotics or DNA synthesis inhibitors/replication stress inducers) have demonstrated success as broad-spectrum anticancer drugs. Cyclin-dependent kinases (CDKs) are drivers of cell cycle transitions. A CDK inhibitor, flavopiridol/alvocidib, is an FDA-approved drug for acute myeloid leukemia. Alzheimer's disease (AD) is another serious issue in contemporary medicine. The cause of AD remains elusive, although a critical role of latent amyloid-beta accumulation has emerged. Existing AD drug research and development targets include amyloid, amyloid metabolism/catabolism, tau, inflammation, cholesterol, the cholinergic system, and other neurotransmitters. However, none have been validated as therapeutically effective targets. Recent reports from AD-omics and preclinical animal models provided data supporting the long-standing notion that cell cycle progression and/or mitosis may be a valid target for AD prevention and/or therapy. This review will summarize the recent developments in AD research: (a) Mitotic re-entry, leading to the "amyloid-beta accumulation cycle," may be a prerequisite for amyloid-beta accumulation and AD pathology development; (b) AD-associated pathogens can cause cell cycle errors; (c) thirteen among 37 human AD genetic risk genes may be functionally involved in the cell cycle and/or mitosis; and (d) preclinical AD mouse models treated with CDK inhibitor showed improvements in cognitive/behavioral symptoms. If the "amyloid-beta accumulation cycle is an AD drug target" concept is proven, repurposing of cancer drugs may emerge as a new, fast-track approach for AD management in the clinic setting.
Collapse
Affiliation(s)
- Chinthalapally V. Rao
- Center for Cancer Prevention and Drug DevelopmentDepartment of MedicineHematology/Oncology SectionUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| | - Adam S. Asch
- Stephenson Cancer CenterDepartment of MedicineHematology/Oncology SectionUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| | - Daniel J. J. Carr
- Department of OphthalmologyUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| | - Hiroshi Y. Yamada
- Center for Cancer Prevention and Drug DevelopmentDepartment of MedicineHematology/Oncology SectionUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| |
Collapse
|
33
|
Jung ME, Metzger DB, Hall J. The long-term but not short-term use of benzodiazepine impairs motoric function and upregulates amyloid β in part through the suppression of translocator protein. Pharmacol Biochem Behav 2020; 191:172873. [PMID: 32105662 DOI: 10.1016/j.pbb.2020.172873] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/22/2020] [Accepted: 02/15/2020] [Indexed: 11/24/2022]
Abstract
Many elderly American women use CNS depressant benzodiazepine (BZD) to ameliorate anxiety or insomnia. However, the chronic use of BZD (cBZD) is prevalent, causing adverse effects of BZD that include movement deficit. We previously reported that cBZD upregulates neurotoxic amyloid β42 (Aβ42) and downregulates neuroprotective translocator protein (TSPO) in the cerebellum, the brain area of movement and balance. The aim of the current study is two-fold: 1) to determine a direct effect of TSPO (inhibition) on cBZD-induced Aβ42 and Aβ-associated molecules; Aβ-producing-protein presenilin-1 (PS1) and Aβ-degrading-enzyme neprilysin and 2) to determine whether Aβ42 upregulation and motoric deficit occur upon a long-term (cBZD) rather than a short-term BZD (sBZD) treatment. Old female mice received BZD (lorazepam) for 20 days (cBZD) or 3 days (sBZD) with or without prototype TSPO ligand PK11195 and were tested for motoric performance for 3 days using Rotarod. ELISA was conducted to measure Aβ42 level and neprilysin activity in cerebellum. RT-PCR and immunoblot were conducted to measure the mRNA and protein levels of TSPO, PS1, and neprilysin. cBZD treatment decreased TSPO and neprilysin but increased Aβ42 accompanied by motoric deficit. Chronic PK11195 treatment acted as a TSPO inhibitor by suppressing TSPO expression and mimicked or exacerbated the effects of cBZD on all parameters measured except for PS1. None of the molecular and behavioral changes induced by cBZD were reproduced by sBZD treatment. These data suggest that cBZD upregulates Aβ42 and downregulates neprilysin in part through TSPO inhibition, the mechanisms distinct from sBZD, collectively contributing to motoric deficit.
Collapse
Affiliation(s)
- Marianna E Jung
- Pharmacology and Neuroscience, UNT Health Science Center, Institute for Healthy Aging, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, United States of America.
| | - Daniel B Metzger
- Pharmacology and Neuroscience, UNT Health Science Center, Institute for Healthy Aging, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, United States of America
| | - James Hall
- Pharmacology and Neuroscience, UNT Health Science Center, Institute for Healthy Aging, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, United States of America
| |
Collapse
|
34
|
Nicastro N, Mak E, Williams GB, Surendranathan A, Bevan-Jones WR, Passamonti L, Vàzquez Rodrìguez P, Su L, Arnold R, Fryer TD, Hong YT, Aigbirhio FI, Rowe JB, O'Brien JT. Correlation of microglial activation with white matter changes in dementia with Lewy bodies. Neuroimage Clin 2020; 25:102200. [PMID: 32032816 PMCID: PMC7005463 DOI: 10.1016/j.nicl.2020.102200] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/26/2020] [Accepted: 01/27/2020] [Indexed: 02/02/2023]
Abstract
Dementia with Lewy bodies (DLB) is characterized by alpha-synuclein protein deposition with variable degree of concurrent Alzheimer's pathology. Neuroinflammation is also increasingly recognized as a significant contributor to degeneration. We aimed to examine the relationship between microglial activation as measured with [11C]-PK11195 brain PET, MR diffusion tensor imaging (DTI) and grey matter atrophy in DLB. Nineteen clinically probable DLB and 20 similarly aged controls underwent 3T structural MRI (T1-weighted) and diffusion-weighted imaging. Eighteen DLB subjects also underwent [11C]-PK11195 PET imaging and 15 had [11C]-Pittsburgh compound B amyloid PET, resulting in 9/15 being amyloid-positive. We used Computational Anatomy Toolbox (CAT12) for volume-based morphometry (VBM) and Tract-Based Spatial Statistics (TBSS) for DTI to assess group comparisons between DLB and controls and to identify associations of [11C]-PK11195 binding with grey/white matter changes and cognitive score in DLB patients. VBM analyses showed that DLB had extensive reduction of grey matter volume in superior frontal, temporal, parietal and occipital cortices (family-wise error (FWE)-corrected p < 0.05). TBSS showed widespread changes in DLB for all DTI parameters (reduced fractional anisotropy, increased diffusivity), involving the corpus callosum, corona radiata and superior longitudinal fasciculus (FWE-corrected p < 0.05). Higher [11C]-PK11195 binding in parietal cortices correlated with widespread lower mean and radial diffusivity in DLB patients (FWE-corrected p < 0.05). Furthermore, preserved cognition in DLB (higher Addenbrookes Cognitive Evaluation revised score) also correlated with higher [11C]-PK11195 binding in frontal, temporal, and occipital lobes. However, microglial activation was not significantly associated with grey matter changes. Our study suggests that increased microglial activation is associated with a relative preservation of white matter and cognition in DLB, positioning neuroinflammation as a potential early marker of DLB etio-pathogenesis.
Collapse
Affiliation(s)
- Nicolas Nicastro
- Department of Psychiatry, University of Cambridge, UK,Department of Clinical Neurosciences, Geneva University Hospitals, Switzerland
| | - Elijah Mak
- Department of Psychiatry, University of Cambridge, UK
| | | | | | | | - Luca Passamonti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK,Consiglio Nazionale delle Ricerche (CNR), Istituto di Bioimmagini e Fisiologia Molecolare (IBFM), Milano, Italy
| | | | - Li Su
- Department of Psychiatry, University of Cambridge, UK,China-UK Centre for Cognition and Ageing Research, Southwest University, Chongqing, China
| | - Robert Arnold
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Tim D. Fryer
- Wolfson Brain Imaging Centre, University of Cambridge, UK,Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Young T. Hong
- Wolfson Brain Imaging Centre, University of Cambridge, UK,Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - James B. Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK,Medical Research Council Cognition and Brain Sciences Unit, Cambridge, UK
| | - John T. O'Brien
- Department of Psychiatry, University of Cambridge, UK,Corresponding author at: Department of Psychiatry, University of Cambridge School of Clinical Medicine, Box 189, Level E4 Cambridge Biomedical Campus, Cambridge CB2 0SP, United Kingdom.
| |
Collapse
|
35
|
Laquinimod ameliorates secondary brain inflammation. Neurobiol Dis 2019; 134:104675. [PMID: 31731041 DOI: 10.1016/j.nbd.2019.104675] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/21/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence suggests that a degenerative processes within the brain can trigger the formation of new, focal inflammatory lesions in Multiple Sclerosis (MS). Here, we used a novel pre-clinical MS animal model to test whether the amelioration of degenerative brain events reduces the secondary recruitment of peripheral immune cells and, in consequence, inflammatory lesion development. Neural degeneration was induced by a 3 weeks cuprizone intoxication period. To mitigate the cuprizone-induced pathology, animals were treated with Laquinimod (25 mg/kg) during the cuprizone-intoxication period. At the beginning of week 6, encephalitogenic T cell development in peripheral lymphoid organs was induced by the immunization with myelin oligodendrocyte glycoprotein 35-55 peptide (i.e., Cup/EAE). Demyelination, axonal injury and reactive gliosis were determined by immunohistochemistry. Positron emission tomography (PET) imaging was performed to analyze glia activation in vivo. Vehicle-treated cuprizone mice displayed extensive callosal demyelination, glia activation and enhanced TSPO-ligand binding. This cuprizone-induced pathology was profoundly ameliorated in mice treated with Laquinimod. In vehicle-treated Cup/EAE mice, the cuprizone-induced pathology triggered massive peripheral immune cell recruitment into the forebrain, evidenced by multifocal perivascular inflammation, glia activation and neuro-axonal injury. While anti myelin oligodendrocyte glycoprotein 35-55 peptide immune responses were comparable in vehicle- and Laquinimod-treated Cup/EAE mice, the cuprizone-triggered immune cell recruitment was ameliorated by the Laquinimod treatment. This study clearly illustrates that amelioration of a primary brain-intrinsic degenerative process secondary halts peripheral immune cell recruitment and, in consequence, inflammatory lesion development. These findings have important consequences for the interpretation of the results of clinical studies.
Collapse
|
36
|
Vermeulen K, Vandamme M, Bormans G, Cleeren F. Design and Challenges of Radiopharmaceuticals. Semin Nucl Med 2019; 49:339-356. [PMID: 31470930 DOI: 10.1053/j.semnuclmed.2019.07.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This review describes general concepts with regard to radiopharmaceuticals for diagnostic or therapeutic applications that help to understand the specific challenges encountered during the design, (radio)synthesis, in vitro and in vivo evaluation and clinical translation of novel radiopharmaceuticals. The design of a radiopharmaceutical requires upfront decisions with regard to combining a suitable vector molecule with an appropriate radionuclide, considering the type and location of the molecular target, the desired application, and the time constraints imposed by the relatively short half-life of radionuclides. Well-designed in vitro and in vivo experiments allow nonclinical validation of radiotracers. Ultimately, in combination with a limited toxicology package, the radiotracer becomes a radiopharmaceutical for clinical evaluation, produced in compliance with regulatory requirements for medicines for intravenous (IV) injection.
Collapse
Affiliation(s)
- Koen Vermeulen
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| | - Mathilde Vandamme
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium.
| | - Frederik Cleeren
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| |
Collapse
|
37
|
Sacher C, Blume T, Beyer L, Peters F, Eckenweber F, Sgobio C, Deussing M, Albert NL, Unterrainer M, Lindner S, Gildehaus FJ, von Ungern-Sternberg B, Brzak I, Neumann U, Saito T, Saido TC, Bartenstein P, Rominger A, Herms J, Brendel M. Longitudinal PET Monitoring of Amyloidosis and Microglial Activation in a Second-Generation Amyloid-β Mouse Model. J Nucl Med 2019; 60:1787-1793. [PMID: 31302633 DOI: 10.2967/jnumed.119.227322] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/15/2019] [Indexed: 11/16/2022] Open
Abstract
Nonphysiologic overexpression of amyloid-β (Aβ) precursor protein in common transgenic Aβ mouse models of Alzheimer disease likely hampers their translational potential. The novel App NL-G-F mouse incorporates a mutated knock-in, potentially presenting an improved model of Alzheimer disease for Aβ-targeting treatment trials. We aimed to establish serial small-animal PET of amyloidosis and neuroinflammation in App NL-G-F mice as a tool for therapy monitoring. Methods: App NL-G-F mice (20 homozygous and 21 heterogeneous) and 12 age-matched wild-type mice were investigated longitudinally from 2.5 to 10 mo of age with 18F-florbetaben Aβ PET and 18F-GE-180 18-kDa translocator protein (TSPO) PET. Voxelwise analysis of SUV ratio images was performed using statistical parametric mapping. All mice underwent a Morris water maze test of spatial learning after their final scan. Quantification of fibrillar Aβ and activated microglia by immunohistochemistry and biochemistry served for validation of the PET results. Results: The periaqueductal gray emerged as a suitable pseudo reference tissue for both tracers. Homozygous App NL-G-F mice had a rising SUV ratio in cortex and hippocampus for Aβ (+9.1%, +3.8%) and TSPO (+19.8%, +14.2%) PET from 2.5 to 10 mo of age (all P < 0.05), whereas heterozygous App NL-G-F mice did not show significant changes with age. Significant voxelwise clusters of Aβ deposition and microglial activation in homozygous mice appeared at 5 mo of age. Immunohistochemical and biochemical findings correlated strongly with the PET data. Water maze escape latency was significantly elevated in homozygous App NL-G-F mice compared with wild-type at 10 mo of age and was associated with high TSPO binding. Conclusion: Longitudinal PET in App NL-G-F knock-in mice enables monitoring of amyloidogenesis and neuroinflammation in homozygous mice but is insensitive to minor changes in heterozygous animals. The combination of PET with behavioral tasks in App NL-G-F treatment trials is poised to provide important insights in preclinical drug development.
Collapse
Affiliation(s)
- Christian Sacher
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich Germany
| | - Tanja Blume
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich Germany.,DZNE-German Center for Neurodegenerative Diseases, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich Germany
| | - Finn Peters
- DZNE-German Center for Neurodegenerative Diseases, Munich, Germany
| | - Florian Eckenweber
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich Germany
| | - Carmelo Sgobio
- DZNE-German Center for Neurodegenerative Diseases, Munich, Germany
| | - Maximilian Deussing
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich Germany
| | - Franz-Josef Gildehaus
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich Germany
| | | | - Irena Brzak
- Neuroscience, Novartis Institutes for BioMedical Research (NIBR), Basel, Switzerland
| | - Ulf Neumann
- Neuroscience, Novartis Institutes for BioMedical Research (NIBR), Basel, Switzerland
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich Germany
| | - Axel Rominger
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland; and
| | - Jochen Herms
- DZNE-German Center for Neurodegenerative Diseases, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Center of Neuropathology and Prion Research, University of Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich Germany .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
38
|
Nack A, Brendel M, Nedelcu J, Daerr M, Nyamoya S, Beyer C, Focke C, Deussing M, Hoornaert C, Ponsaerts P, Schmitz C, Bartenstein P, Rominger A, Kipp M. Expression of Translocator Protein and [18F]-GE180 Ligand Uptake in Multiple Sclerosis Animal Models. Cells 2019; 8:cells8020094. [PMID: 30696113 PMCID: PMC6406715 DOI: 10.3390/cells8020094] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/16/2019] [Accepted: 01/23/2019] [Indexed: 12/19/2022] Open
Abstract
Positron emission tomography (PET) ligands targeting the translocator protein (TSPO) represent promising tools to visualize neuroinflammation in multiple sclerosis (MS). Although it is known that TSPO is expressed in the outer mitochondria membrane, its cellular localization in the central nervous system under physiological and pathological conditions is not entirely clear. The purpose of this study was to assess the feasibility of utilizing PET imaging with the TSPO tracer, [18F]-GE180, to detect histopathological changes during experimental demyelination, and to determine which cell types express TSPO. C57BL/6 mice were fed with cuprizone for up to 5 weeks to induce demyelination. Groups of mice were investigated by [18F]-GE180 PET imaging at week 5. Recruitment of peripheral immune cells was triggered by combining cuprizone intoxication with MOG35–55 immunization (i.e., Cup/EAE). Immunofluorescence double-labelling and transgene mice were used to determine which cell types express TSPO. [18F]-GE180-PET reliably detected the cuprizone-induced pathology in various white and grey matter regions, including the corpus callosum, cortex, hippocampus, thalamus and caudoputamen. Cuprizone-induced demyelination was paralleled by an increase in TSPO expression, glia activation and axonal injury. Most of the microglia and around one-third of the astrocytes expressed TSPO. TSPO expression induction was more severe in the white matter corpus callosum compared to the grey matter cortex. Although mitochondria accumulate at sites of focal axonal injury, these mitochondria do not express TSPO. In Cup/EAE mice, both microglia and recruited monocytes contribute to the TSPO expressing cell populations. These findings support the notion that TSPO is a valuable marker for the in vivo visualization and quantification of neuropathological changes in the MS brain. The pathological substrate of an increase in TSPO-ligand binding might be diverse including microglia activation, peripheral monocyte recruitment, or astrocytosis, but not axonal injury.
Collapse
MESH Headings
- Animals
- Astrocytes/pathology
- Astrocytes/ultrastructure
- Axons/metabolism
- Axons/ultrastructure
- Biomarkers/metabolism
- Carbazoles/metabolism
- Cuprizone
- Demyelinating Diseases/diagnostic imaging
- Demyelinating Diseases/pathology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/diagnostic imaging
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Inflammation/pathology
- Ligands
- Mice, Inbred C57BL
- Mitochondria/metabolism
- Mitochondria/ultrastructure
- Monocytes/metabolism
- Multiple Sclerosis/diagnostic imaging
- Neuroglia/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, GABA/genetics
- Receptors, GABA/metabolism
Collapse
Affiliation(s)
- Anne Nack
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany.
- Department of Anatomy, 39071 Rostock University Medical Center, Rostock, Germany.
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, 80336 Munich, Germany.
| | - Julia Nedelcu
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany.
- Department of Anatomy, 39071 Rostock University Medical Center, Rostock, Germany.
| | - Markus Daerr
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany.
- Department of Anatomy, 39071 Rostock University Medical Center, Rostock, Germany.
| | - Stella Nyamoya
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany.
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany.
- Department of Anatomy, 39071 Rostock University Medical Center, Rostock, Germany.
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany.
| | - Carola Focke
- Department of Nuclear Medicine, University Hospital, LMU Munich, 80336 Munich, Germany.
| | - Maximilian Deussing
- Department of Nuclear Medicine, University Hospital, LMU Munich, 80336 Munich, Germany.
| | - Chloé Hoornaert
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium.
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium.
| | - Christoph Schmitz
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany.
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, 80336 Munich, Germany.
| | - Axel Rominger
- Department of Nuclear Medicine, University Hospital, LMU Munich, 80336 Munich, Germany.
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland.
| | - Markus Kipp
- Department of Anatomy, 39071 Rostock University Medical Center, Rostock, Germany.
| |
Collapse
|