1
|
Wang Y, Liao W, Ye K, Wang Y, Li J, Huang D, Hong X, Cheng W, Luan P, Tian J. Correlation between Olfaction and Cognition Using PET Quantification of Alzheimer's Disease Biomarkers in Intracranial Olfactory-Related Regions of 3xTg Mice. Mol Pharm 2025. [PMID: 40235036 DOI: 10.1021/acs.molpharmaceut.4c01532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Olfactory impairment is a preclinical symptom of Alzheimer's disease (AD); however, the mechanism of olfactory dysfunction in AD is not clear. This study aims to explore the mechanism of olfactory dysfunction in AD by analyzing the correlation between olfaction and cognition using positron emission tomography (PET) quantification of biomarkers, including amyloid β (Aβ) and abnormal tau proteins in the intracranial olfactory-related regions of AD model mice, and compares PET quantification with pathological detection to explore the reliability of the transformation of PET into clinical research. A total of 14 mice, including 3xTg mice (12 months old, n = 7) and wild-type (WT) mice (12 months old, n = 7), were enrolled in the study. Behavioral experiments (olfactory: buried food test, cognitive: Morris water maze) were performed to assess whether the mice had abnormal olfactory and cognitive functions. PET quantified Aβ and abnormal tau in olfactory-related regions. The expression and distribution of Aβ and phosphorylated tau were observed by immunofluorescence. Compared with WT mice, 3xTg mice had olfactory and cognitive impairments. Quantitative PET and immunofluorescence showed that 3xTg mice had significantly more Aβ and abnormal tau accumulation in olfactory areas than did WT mice. The deposition of Aβ and phosphorylated tau in olfactory-related regions of 3xTg mice suggests a potential link between olfactory pathology and AD progression. The elevated uptake of Aβ and tau PET in the olfactory-related regions further highlights the potential of olfactory-targeted molecular imaging as a noninvasive biomarker for AD detection.
Collapse
Affiliation(s)
- Yulin Wang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
| | - Wanchen Liao
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
- School of Basic Medical Sciences, Shenzhen University, Shenzhen 518060, China
| | - Kaiyu Ye
- School of Basic Medical Sciences, Shenzhen University, Shenzhen 518060, China
| | - Yuqi Wang
- School of Basic Medical Sciences, Shenzhen University, Shenzhen 518060, China
| | - Jun Li
- School of Basic Medical Sciences, Shenzhen University, Shenzhen 518060, China
| | - Dongqing Huang
- School of Basic Medical Sciences, Shenzhen University, Shenzhen 518060, China
| | - Xinyang Hong
- School of Basic Medical Sciences, Shenzhen University, Shenzhen 518060, China
| | - Weibin Cheng
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
| | - Ping Luan
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
- School of Basic Medical Sciences, Shenzhen University, Shenzhen 518060, China
| | - Junzhang Tian
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
| |
Collapse
|
2
|
Pierson SR, Fiock KL, Wang R, Balasubramanian N, Reinhardt J, Khan KM, James TD, Hunter ML, Cooper BJ, Williamsen HR, Betters R, Deniz K, Lee G, Aldridge G, Hefti MM, Marcinkiewcz CA. Tau pathology in the dorsal raphe may be a prodromal indicator of Alzheimer's disease. Mol Psychiatry 2025; 30:532-546. [PMID: 39143322 PMCID: PMC12010729 DOI: 10.1038/s41380-024-02664-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 06/22/2024] [Accepted: 07/04/2024] [Indexed: 08/16/2024]
Abstract
Protein aggregation in brainstem nuclei is thought to occur in the early stages of Alzheimer's disease (AD), but its specific role in driving prodromal symptoms and disease progression is largely unknown. The dorsal raphe nucleus (DRN) contains a large population of serotonin (5-hydroxytryptamine; 5-HT) neurons that regulate mood, reward-related behavior, and sleep, which are all disrupted in AD. We report here that tau pathology is present in the DRN of individuals 25-80 years old without a known history of dementia, and its prevalence was comparable to the locus coeruleus (LC). By comparison, fewer cases were positive for other pathological proteins including α-synuclein, β-amyloid, and TDP-43. To evaluate how early tau pathology impacts behavior, we overexpressed human P301L-tau in the DRN of mice and observed depressive-like behaviors and hyperactivity without deficits in spatial memory. Tau pathology was predominantly found in neurons relative to glia and colocalized with a significant proportion of Tph2-expressing neurons in the DRN. 5-HT neurons were also hyperexcitable in P301L-tauDRN mice, and there was an increase in the amplitude of excitatory post-synaptic currents (EPSCs). Moreover, astrocytic density was elevated in the DRN and accompanied by an increase in IL-1α and Frk expression, which suggests increased inflammatory signaling. Additionally, tau pathology was detected in axonal processes in the thalamus, hypothalamus, amygdala, and caudate putamen. A significant proportion of this tau pathology colocalized with the serotonin reuptake transporter (SERT), suggesting that tau may spread in an anterograde manner to regions outside the DRN. Together these results indicate that tau pathology accumulates in the DRN in a subset of individuals over 50 years and may lead to behavioral dysregulation, 5-HT neuronal dysfunction, and activation of local astrocytes which may be prodromal indicators of AD.
Collapse
Affiliation(s)
- Samantha R Pierson
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA
| | - Kimberly L Fiock
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | - Ruixiang Wang
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA
| | | | - Jessica Reinhardt
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA
| | - Kanza M Khan
- Psychological Sciences Department, Daemen University, Amherst, NY, 14226, USA
| | - Thomas D James
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA
| | - Mikayla L Hunter
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | - Benjamin J Cooper
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | | | - Ryan Betters
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | - Kaancan Deniz
- Department of Neurology, University of Iowa, Iowa City, IA, 52242, USA
| | - Gloria Lee
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| | - Georgina Aldridge
- Department of Neurology, University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| | - Marco M Hefti
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| | - Catherine A Marcinkiewcz
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
3
|
Ramakrishnan NK, Zhao AZ, Thompson S, Milicevic Sephton S, Williamson DJ, Smolek T, Žilka N, Aigbirhio FI. PET Imaging of a Transgenic Tau Rat Model SHR24 with [ 18F]AV1451. Mol Imaging Biol 2025:10.1007/s11307-024-01972-4. [PMID: 39838233 DOI: 10.1007/s11307-024-01972-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/30/2024] [Accepted: 11/25/2024] [Indexed: 01/23/2025]
Abstract
PURPOSE Positron Emission Tomography (PET) scans with radioligands targeting tau neurofibrillary tangles (NFT) have accelerated our understanding of the role of misfolded tau in neurodegeneration. While intended for human research, applying these radioligands to small animals establishes a vital translational link. Transgenic animal models of dementia, such as the tau rat SHR24, play a crucial role in enhancing our understanding of these disorders. This study aims to evaluate the utility of SHR24 rat model for PET studies. PROCEDURES Dynamic PET scans were conducted in male SHR24 rats and their wild-type SHR (SHRwt) littermates using [18F]AV1451. Rapid blood sampling and metabolite analysis were performed to acquire input curves. Time activity curves were obtained from various brain regions over 60 min. Blood-based, 2-Tissue Compartment Model (2-TCM) and Logan graphical analysis were used to obtain kinetic modelling parameters. The ability of reference tissue models to predict the binding potential (BPND) were assessed. Autoradiography studies were performed to corroborate the scan data. RESULTS Total distribution volume (VT) was the best predicted parameter which revealed significantly higher uptake of [18F]AV1451 in the cortex (5.8 ± 1.1 vs 4.6 ± 0.7, P < 0.05) of SHR24 rats compared to SHRwt rats. Binding potential obtained from 2-TCM was variable, however BPND from reference tissue models detected significantly higher binding in cortex (0.28 ± 0.07 vs 0.20 ± 0.04, P < 0.01 by SRTM) and brainstem (0.14 ± 0.04 vs 0.08 ± 0.02, P < 0.01, by SRTM). CONCLUSIONS With the ability to detect binding of established radioligand [18F]AV1451 in these rats, we have demonstrated the utility of this model for assessing aggregated tau neurobiology by PET, with reference tissue models providing potential for longitudinal studies.
Collapse
Affiliation(s)
- Nisha K Ramakrishnan
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK.
- Preclinical Imaging Research Laboratory, Anne McLaren Building, 90 Francis Crick Avenue, Trumpington, Cambridge, CB2 0BA, UK.
| | - Annie Ziyi Zhao
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Stephen Thompson
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Selena Milicevic Sephton
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - David J Williamson
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Tomáš Smolek
- Axon Neuroscience R&D Services SE, Dubravska vćesta 9, 811 02, Bratislava, Slovakia
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10, Bratislava, Slovakia
| | - Norbert Žilka
- Axon Neuroscience R&D Services SE, Dubravska vćesta 9, 811 02, Bratislava, Slovakia
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10, Bratislava, Slovakia
| | - Franklin I Aigbirhio
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
| |
Collapse
|
4
|
Slemann L, Gnörich J, Hummel S, Bartos LM, Klaus C, Kling A, Kusche-Palenga J, Kunte ST, Kunze LH, Englert AL, Li Y, Vogler L, Katzdobler S, Palleis C, Bernhardt A, Jäck A, Zwergal A, Hopfner F, Roemer-Cassiano SN, Biechele G, Stöcklein S, Bischof G, van Eimeren T, Drzezga A, Sabri O, Barthel H, Respondek G, Grimmer T, Levin J, Herms J, Paeger L, Willroider M, Beyer L, Höglinger GU, Roeber S, Franzmeier N, Brendel M. Neuronal and oligodendroglial, but not astroglial, tau translates to in vivo tau PET signals in individuals with primary tauopathies. Acta Neuropathol 2024; 148:70. [PMID: 39580770 PMCID: PMC11586312 DOI: 10.1007/s00401-024-02834-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/14/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024]
Abstract
Tau PET has attracted increasing interest as an imaging biomarker for 4-repeat (4R)-tauopathy progressive supranuclear palsy (PSP). However, the translation of in vitro 4R-tau binding to in vivo tau PET signals is still unclear. Therefore, we performed a translational study using a broad spectrum of advanced methodologies to investigate the sources of [18F]PI-2620 tau PET signals in individuals with 4R-tauopathies, including a pilot PET autopsy study in patients. First, we conducted a longitudinal [18F]PI-2620 PET/MRI study in a 4-repeat-tau mouse model (PS19) and detected elevated [18F]PI-2620 PET signals in the presence of high levels of neuronal tau. An innovative approach involving cell sorting after radiotracer injection in vivo revealed higher tracer uptake in single neurons than in the astrocytes of PS19 mice. Regional [18F]PI-2620 tau PET signals during the lifetime correlated with the abundance of fibrillary tau and with autoradiography signal intensity in PSP patients and disease controls who underwent autopsy 2-63 months after tau PET. In autoradiography, tau-positive neurons and oligodendrocytes with a high AT8 density, but not tau-positive astrocytes, were the drivers of [18F]PI-2620 autoradiography signals in individuals with PSP. The high tau abundance in oligodendrocytes at the boundary of gray and white matter facilitated the identification of an optimized frontal lobe target region to detect the tau burden in patients with PSP. In summary, neuronal and oligodendroglial tau constitutes the dominant source of tau PET radiotracer binding in 4-repeat-tauopathies, translating to an in vivo signal.
Collapse
Affiliation(s)
- Luna Slemann
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Johannes Gnörich
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Selina Hummel
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Laura M Bartos
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Carolin Klaus
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Agnes Kling
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Julia Kusche-Palenga
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Sebastian T Kunte
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Lea H Kunze
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Amelie L Englert
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Yunlei Li
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Letizia Vogler
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Sabrina Katzdobler
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Carla Palleis
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Alexander Bernhardt
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Alexander Jäck
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Andreas Zwergal
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- German Center for Vertigo and Balance Disorders, DSGZ, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Franziska Hopfner
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Sebastian N Roemer-Cassiano
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Institute for Stroke and Dementia Research, LMU Hospital, LMU Munich, Munich, Germany
| | - Gloria Biechele
- Department of Radiology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Sophia Stöcklein
- Department of Radiology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Gerard Bischof
- Cognitive Neuroscience, Institute for Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| | - Thilo van Eimeren
- Cognitive Neuroscience, Institute for Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
- Department of Neurology, University Hospital Cologne, Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Gesine Respondek
- Department of Neurology, Medizinische Hochschule Hannover, Hannover, Germany
| | - Timo Grimmer
- Center for Cognitive Disorders, Department of Psychiatry and Psychotherapy, School of Medicine and Health, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Center of Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Lars Paeger
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Marie Willroider
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Sigrun Roeber
- Center of Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Nicolai Franzmeier
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute for Stroke and Dementia Research, LMU Hospital, LMU Munich, Munich, Germany
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, , University of Gothenburg, The Sahlgrenska Academy, Mölndal, Gothenburg, Sweden
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
5
|
Bartos LM, Quach S, Zenatti V, Kirchleitner SV, Blobner J, Wind-Mark K, Kolabas ZI, Ulukaya S, Holzgreve A, Ruf VC, Kunze LH, Kunte ST, Hoermann L, Härtel M, Park HE, Groß M, Franzmeier N, Zatcepin A, Zounek A, Kaiser L, Riemenschneider MJ, Perneczky R, Rauchmann BS, Stöcklein S, Ziegler S, Herms J, Ertürk A, Tonn JC, Thon N, von Baumgarten L, Prestel M, Tahirovic S, Albert NL, Brendel M. Remote Neuroinflammation in Newly Diagnosed Glioblastoma Correlates with Unfavorable Clinical Outcome. Clin Cancer Res 2024; 30:4618-4634. [PMID: 39150564 PMCID: PMC11474166 DOI: 10.1158/1078-0432.ccr-24-1563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/15/2024] [Accepted: 08/14/2024] [Indexed: 08/17/2024]
Abstract
PURPOSE Current therapy strategies still provide only limited success in the treatment of glioblastoma, the most frequent primary brain tumor in adults. In addition to the characterization of the tumor microenvironment, global changes in the brain of patients with glioblastoma have been described. However, the impact and molecular signature of neuroinflammation distant of the primary tumor site have not yet been thoroughly elucidated. EXPERIMENTAL DESIGN We performed translocator protein (TSPO)-PET in patients with newly diagnosed glioblastoma (n = 41), astrocytoma WHO grade 2 (n = 7), and healthy controls (n = 20) and compared TSPO-PET signals of the non-lesion (i.e., contralateral) hemisphere. Back-translation into syngeneic SB28 glioblastoma mice was used to characterize Pet alterations on a cellular level. Ultimately, multiplex gene expression analyses served to profile immune cells in remote brain. RESULTS Our study revealed elevated TSPO-PET signals in contralateral hemispheres of patients with newly diagnosed glioblastoma compared to healthy controls. Contralateral TSPO was associated with persisting epileptic seizures and shorter overall survival independent of the tumor phenotype. Back-translation into syngeneic glioblastoma mice pinpointed myeloid cells as the predominant source of contralateral TSPO-PET signal increases and identified a complex immune signature characterized by myeloid cell activation and immunosuppression in distant brain regions. CONCLUSIONS Neuroinflammation within the contralateral hemisphere can be detected with TSPO-PET imaging and associates with poor outcome in patients with newly diagnosed glioblastoma. The molecular signature of remote neuroinflammation promotes the evaluation of immunomodulatory strategies in patients with detrimental whole brain inflammation as reflected by high TSPO expression.
Collapse
Affiliation(s)
- Laura M. Bartos
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Stefanie Quach
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany.
| | - Valerio Zenatti
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
| | | | - Jens Blobner
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany.
| | - Karin Wind-Mark
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Zeynep Ilgin Kolabas
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Munich, Germany.
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
- Graduate School of Systemic Neurosciences (GSN), Munich, Germany.
| | - Selin Ulukaya
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Munich, Germany.
- Faculty of Biology, Master of Science Program in Molecular and Cellular Biology, Ludwig-Maximilians-Universität München, Planegg, Germany.
| | - Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Viktoria C. Ruf
- Center for Neuropathology and Prion Research, University Hospital, LMU Munich, Munich, Germany.
| | - Lea H. Kunze
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Sebastian T. Kunte
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Leonie Hoermann
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Marlies Härtel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Ha Eun Park
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Mattes Groß
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
| | - Artem Zatcepin
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
| | - Adrian Zounek
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Lena Kaiser
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | | | - Robert Perneczky
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), University of Munich, Munich, Germany.
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany.
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom.
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, United Kingdom.
| | | | - Sophia Stöcklein
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany.
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Jochen Herms
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
- Center for Neuropathology and Prion Research, University Hospital, LMU Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), University of Munich, Munich, Germany.
| | - Ali Ertürk
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Munich, Germany.
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
- Graduate School of Systemic Neurosciences (GSN), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), University of Munich, Munich, Germany.
| | - Joerg C. Tonn
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| | - Niklas Thon
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Louisa von Baumgarten
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| | - Matthias Prestel
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
| | - Sabina Tahirovic
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
| | - Nathalie L. Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Munich Cluster for Systems Neurology (SyNergy), University of Munich, Munich, Germany.
| |
Collapse
|
6
|
Liang M, Gu L, Zhang H, Min J, Wang Z, Ma Z, Zhang C, Zeng S, Pan Y, Yan D, Shen Z, Huang W. Design, Synthesis, and Bioactivity of Novel Bifunctional Small Molecules for Alzheimer's disease. ACS OMEGA 2022; 7:26308-26315. [PMID: 35936449 PMCID: PMC9352321 DOI: 10.1021/acsomega.2c02130] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The abnormal phosphorylation of the τ-protein is a typical early pathological feature of Alzheimer's disease (AD). The excessive phosphorylation of the τ-protein in the brain causes the formation of neurofibrillary tangles (NFTs) and increases the neurotoxicity of amyloid-β (Aβ). Thus, targeting the τ-protein is considered a promising strategy for treating AD. Herein, we designed and synthesized a series of molecules containing bifunctional groups to recognize the τ-protein and the E3 ligase. The molecules were examined in vitro, and their effects were tested on PC12 cells. In addition, we further studied the pharmacokinetics of compound I3 in healthy rats. Our data showed that compound I3 could effectively degrade τ-protein, reduce Aβ-induced cytotoxicity, and regulate the uneven distribution of mitochondria, which may open a new therapeutic strategy for the treatment of AD.
Collapse
Affiliation(s)
- Meihao Liang
- Affiliated
Yongkang First People’s Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, China
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Lili Gu
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Hongjie Zhang
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Jingli Min
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Zunyuan Wang
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Zhen Ma
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Chixiao Zhang
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Shenxin Zeng
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Youlu Pan
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Dongmei Yan
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Zhengrong Shen
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Wenhai Huang
- Affiliated
Yongkang First People’s Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, China
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| |
Collapse
|
7
|
Kimura T, Ono M, Seki C, Sampei K, Shimojo M, Kawamura K, Zhang MR, Sahara N, Takado Y, Higuchi M. A quantitative in vivo imaging platform for tracking pathological tau depositions and resultant neuronal death in a mouse model. Eur J Nucl Med Mol Imaging 2022; 49:4298-4311. [PMID: 35798978 DOI: 10.1007/s00259-022-05898-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 06/28/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE Depositions of tau fibrils are implicated in diverse neurodegenerative disorders, including Alzheimer's disease, and precise assessments of tau pathologies and their impacts on neuronal survival are crucial for pursuing the neurodegenerative tau pathogenesis with and without potential therapies. We aimed to establish an in vivo imaging system to quantify tau accumulations with positron emission tomography (PET) and brain atrophy with volumetric MRI in rTg4510 transgenic mice modeling neurodegenerative tauopathies. METHODS A total of 91 rTg4510 and non-transgenic control mice underwent PET with a tau radiotracer, 18F-PM-PBB3, and MRI at various ages (1.8-12.3 months). Using the cerebellum as reference, the radiotracer binding in target regions was estimated as standardized uptake value ratio (SUVR) and distribution volume ratio (DVR). Histopathological staining of brain sections derived from scanned animals was also conducted to investigate the imaging-neuropathology correlations. RESULTS 18F-PM-PBB3 SUVR at 40-60 min in the neocortex, hippocampus, and striatum of rTg4510 mice agreed with DVR, became significantly different from control values around 4-5 months of age, and progressively and negatively correlated with age and local volumes, respectively. Neocortical SUVR also correlated with the abundance of tau inclusions labeled with PM-PBB3 fluorescence, Gallyas-Braak silver impregnation, and anti-phospho-tau antibodies in postmortem assays. The in vivo and ex vivo 18F-PM-PBB3 binding was blocked by non-radioactive PM-PBB3. 18F-PM-PBB3 yielded a 1.6-fold greater dynamic range for tau imaging than its ancestor, 11C-PBB3. CONCLUSION Our imaging platform has enabled the quantification of tau depositions and consequent neuronal loss and is potentially applicable to the evaluation of candidate anti-tau and neuroprotective drugs.
Collapse
Affiliation(s)
- Taeko Kimura
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Maiko Ono
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Chie Seki
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan.
| | - Kazuaki Sampei
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Masafumi Shimojo
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Kazunori Kawamura
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Ming-Rong Zhang
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Naruhiko Sahara
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Yuhei Takado
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan.
| | - Makoto Higuchi
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| |
Collapse
|
8
|
Vagenknecht P, Luzgin A, Ono M, Ji B, Higuchi M, Noain D, Maschio CA, Sobek J, Chen Z, Konietzko U, Gerez JA, Riek R, Razansky D, Klohs J, Nitsch RM, Dean-Ben XL, Ni R. Non-invasive imaging of tau-targeted probe uptake by whole brain multi-spectral optoacoustic tomography. Eur J Nucl Med Mol Imaging 2022; 49:2137-2152. [PMID: 35128565 PMCID: PMC9165274 DOI: 10.1007/s00259-022-05708-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/25/2022] [Indexed: 02/06/2023]
Abstract
PURPOSE Abnormal tau accumulation within the brain plays an important role in tauopathies such as Alzheimer's disease and frontotemporal dementia. High-resolution imaging of tau deposits at the whole-brain scale in animal disease models is highly desired. METHODS We approached this challenge by non-invasively imaging the brains of P301L mice of 4-repeat tau with concurrent volumetric multi-spectral optoacoustic tomography (vMSOT) at ~ 115 μm spatial resolution using the tau-targeted pyridinyl-butadienyl-benzothiazole derivative PBB5 (i.v.). In vitro probe characterization, concurrent vMSOT and epi-fluorescence imaging of in vivo PBB5 targeting (i.v.) was performed in P301L and wild-type mice, followed by ex vivo validation using AT-8 antibody for phosphorylated tau. RESULTS PBB5 showed specific binding to recombinant K18 tau fibrils by fluorescence assay, to post-mortem Alzheimer's disease brain tissue homogenate by competitive binding against [11C]PBB3 and to tau deposits (AT-8 positive) in post-mortem corticobasal degeneration and progressive supranuclear palsy brains. Dose-dependent optoacoustic and fluorescence signal intensities were observed in the mouse brains following i.v. administration of different concentrations of PBB5. In vivo vMSOT brain imaging of P301L mice showed higher retention of PBB5 in the tau-laden cortex and hippocampus compared to wild-type mice, as confirmed by ex vivo vMSOT, epi-fluorescence, multiphoton microscopy, and immunofluorescence staining. CONCLUSIONS We demonstrated non-invasive whole-brain imaging of tau in P301L mice with vMSOT system using PBB5 at a previously unachieved ~ 115 μm spatial resolution. This platform provides a new tool to study tau spreading and clearance in a tauopathy mouse model, foreseeable in monitoring tau targeting putative therapeutics.
Collapse
Affiliation(s)
- Patrick Vagenknecht
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Artur Luzgin
- Zentrum für Neurowissenschaften Zürich (ZNZ), Zurich, Switzerland
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, ETH Zurich & University of Zurich, Zurich, Switzerland
| | - Maiko Ono
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Bin Ji
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- Department of Radiopharmacy and Molecular Imaging, School of Pharmacy, Fudan University, Shanghai, China
| | - Makoto Higuchi
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Daniela Noain
- Neurology Department, University Hospital Zurich, Zurich, Switzerland
| | - Cinzia A Maschio
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Zentrum für Neurowissenschaften Zürich (ZNZ), Zurich, Switzerland
| | - Jens Sobek
- Functional Genomics Center, University of Zurich, Zurich, Switzerland
| | - Zhenyue Chen
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, ETH Zurich & University of Zurich, Zurich, Switzerland
| | - Uwe Konietzko
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Juan A Gerez
- Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Roland Riek
- Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Daniel Razansky
- Zentrum für Neurowissenschaften Zürich (ZNZ), Zurich, Switzerland
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, ETH Zurich & University of Zurich, Zurich, Switzerland
| | - Jan Klohs
- Zentrum für Neurowissenschaften Zürich (ZNZ), Zurich, Switzerland
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, ETH Zurich & University of Zurich, Zurich, Switzerland
| | - Roger M Nitsch
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Zentrum für Neurowissenschaften Zürich (ZNZ), Zurich, Switzerland
| | - Xose Luis Dean-Ben
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, ETH Zurich & University of Zurich, Zurich, Switzerland.
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.
- Zentrum für Neurowissenschaften Zürich (ZNZ), Zurich, Switzerland.
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, ETH Zurich & University of Zurich, Zurich, Switzerland.
| |
Collapse
|
9
|
Chen B, Marquez-Nostra B, Belitzky E, Toyonaga T, Tong J, Huang Y, Cai Z. PET Imaging in Animal Models of Alzheimer’s Disease. Front Neurosci 2022; 16:872509. [PMID: 35685772 PMCID: PMC9171374 DOI: 10.3389/fnins.2022.872509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
The successful development and translation of PET imaging agents targeting β-amyloid plaques and hyperphosphorylated tau tangles have allowed for in vivo detection of these hallmarks of Alzheimer’s disease (AD) antemortem. Amyloid and tau PET have been incorporated into the A/T/N scheme for AD characterization and have become an integral part of ongoing clinical trials to screen patients for enrollment, prove drug action mechanisms, and monitor therapeutic effects. Meanwhile, preclinical PET imaging in animal models of AD can provide supportive information for mechanistic studies. With the recent advancement of gene editing technologies and AD animal model development, preclinical PET imaging in AD models will further facilitate our understanding of AD pathogenesis/progression and the development of novel treatments. In this study, we review the current state-of-the-art in preclinical PET imaging using animal models of AD and suggest future research directions.
Collapse
|
10
|
Holzgreve A, Pötter D, Brendel M, Orth M, Weidner L, Gold L, Kirchner MA, Bartos LM, Unterrainer LM, Unterrainer M, Steiger K, von Baumgarten L, Niyazi M, Belka C, Bartenstein P, Riemenschneider MJ, Lauber K, Albert NL. Longitudinal [ 18F]GE-180 PET Imaging Facilitates In Vivo Monitoring of TSPO Expression in the GL261 Glioblastoma Mouse Model. Biomedicines 2022; 10:biomedicines10040738. [PMID: 35453488 PMCID: PMC9030822 DOI: 10.3390/biomedicines10040738] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 02/01/2023] Open
Abstract
The 18 kDa translocator protein (TSPO) is increasingly recognized as an interesting target for the imaging of glioblastoma (GBM). Here, we investigated TSPO PET imaging and autoradiography in the frequently used GL261 glioblastoma mouse model and aimed to generate insights into the temporal evolution of TSPO radioligand uptake in glioblastoma in a preclinical setting. We performed a longitudinal [18F]GE-180 PET imaging study from day 4 to 14 post inoculation in the orthotopic syngeneic GL261 GBM mouse model (n = 21 GBM mice, n = 3 sham mice). Contrast-enhanced computed tomography (CT) was performed at the day of the final PET scan (±1 day). [18F]GE-180 autoradiography was performed on day 7, 11 and 14 (ex vivo: n = 13 GBM mice, n = 1 sham mouse; in vitro: n = 21 GBM mice; n = 2 sham mice). Brain sections were also used for hematoxylin and eosin (H&E) staining and TSPO immunohistochemistry. [18F]GE-180 uptake in PET was elevated at the site of inoculation in GBM mice as compared to sham mice at day 11 and later (at day 14, TBRmax +27% compared to sham mice, p = 0.001). In GBM mice, [18F]GE-180 uptake continuously increased over time, e.g., at day 11, mean TBRmax +16% compared to day 4, p = 0.011. [18F]GE-180 uptake as depicted by PET was in all mice co-localized with contrast-enhancement in CT and tissue-based findings. [18F]GE-180 ex vivo and in vitro autoradiography showed highly congruent tracer distribution (r = 0.99, n = 13, p < 0.001). In conclusion, [18F]GE-180 PET imaging facilitates non-invasive in vivo monitoring of TSPO expression in the GL261 GBM mouse model. [18F]GE-180 in vitro autoradiography is a convenient surrogate for ex vivo autoradiography, allowing for straightforward identification of suitable models and scan time-points on previously generated tissue sections.
Collapse
Affiliation(s)
- Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; (A.H.); (D.P.); (M.B.); (L.G.); (M.A.K.); (L.M.B.); (L.M.U.); (P.B.)
| | - Dennis Pötter
- Department of Nuclear Medicine, University Hospital, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; (A.H.); (D.P.); (M.B.); (L.G.); (M.A.K.); (L.M.B.); (L.M.U.); (P.B.)
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; (A.H.); (D.P.); (M.B.); (L.G.); (M.A.K.); (L.M.B.); (L.M.U.); (P.B.)
| | - Michael Orth
- Department of Radiation Oncology, University Hospital, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; (M.O.); (M.N.); (C.B.); (K.L.)
| | - Lorraine Weidner
- Department of Neuropathology, Regensburg University Hospital, 93053 Regensburg, Germany; (L.W.); (M.J.R.)
| | - Lukas Gold
- Department of Nuclear Medicine, University Hospital, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; (A.H.); (D.P.); (M.B.); (L.G.); (M.A.K.); (L.M.B.); (L.M.U.); (P.B.)
| | - Maximilian A. Kirchner
- Department of Nuclear Medicine, University Hospital, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; (A.H.); (D.P.); (M.B.); (L.G.); (M.A.K.); (L.M.B.); (L.M.U.); (P.B.)
| | - Laura M. Bartos
- Department of Nuclear Medicine, University Hospital, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; (A.H.); (D.P.); (M.B.); (L.G.); (M.A.K.); (L.M.B.); (L.M.U.); (P.B.)
| | - Lena M. Unterrainer
- Department of Nuclear Medicine, University Hospital, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; (A.H.); (D.P.); (M.B.); (L.G.); (M.A.K.); (L.M.B.); (L.M.U.); (P.B.)
| | - Marcus Unterrainer
- Department of Radiology, University Hospital, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany;
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (K.S.); (L.v.B.)
| | - Katja Steiger
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (K.S.); (L.v.B.)
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Louisa von Baumgarten
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (K.S.); (L.v.B.)
- Department of Neurosurgery, University Hospital, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany
| | - Maximilian Niyazi
- Department of Radiation Oncology, University Hospital, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; (M.O.); (M.N.); (C.B.); (K.L.)
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (K.S.); (L.v.B.)
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; (M.O.); (M.N.); (C.B.); (K.L.)
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (K.S.); (L.v.B.)
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; (A.H.); (D.P.); (M.B.); (L.G.); (M.A.K.); (L.M.B.); (L.M.U.); (P.B.)
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (K.S.); (L.v.B.)
| | - Markus J. Riemenschneider
- Department of Neuropathology, Regensburg University Hospital, 93053 Regensburg, Germany; (L.W.); (M.J.R.)
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; (M.O.); (M.N.); (C.B.); (K.L.)
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (K.S.); (L.v.B.)
| | - Nathalie L. Albert
- Department of Nuclear Medicine, University Hospital, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; (A.H.); (D.P.); (M.B.); (L.G.); (M.A.K.); (L.M.B.); (L.M.U.); (P.B.)
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (K.S.); (L.v.B.)
- Correspondence:
| |
Collapse
|
11
|
Neuroimaging of Mouse Models of Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10020305. [PMID: 35203515 PMCID: PMC8869427 DOI: 10.3390/biomedicines10020305] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 12/23/2022] Open
Abstract
Magnetic resonance imaging (MRI) and positron emission tomography (PET) have made great strides in the diagnosis and our understanding of Alzheimer’s Disease (AD). Despite the knowledge gained from human studies, mouse models have and continue to play an important role in deciphering the cellular and molecular evolution of AD. MRI and PET are now being increasingly used to investigate neuroimaging features in mouse models and provide the basis for rapid translation to the clinical setting. Here, we provide an overview of the human MRI and PET imaging landscape as a prelude to an in-depth review of preclinical imaging in mice. A broad range of mouse models recapitulate certain aspects of the human AD, but no single model simulates the human disease spectrum. We focused on the two of the most popular mouse models, the 3xTg-AD and the 5xFAD models, and we summarized all known published MRI and PET imaging data, including contrasting findings. The goal of this review is to provide the reader with broad framework to guide future studies in existing and future mouse models of AD. We also highlight aspects of MRI and PET imaging that could be improved to increase rigor and reproducibility in future imaging studies.
Collapse
|
12
|
Ni R. Magnetic Resonance Imaging in Tauopathy Animal Models. Front Aging Neurosci 2022; 13:791679. [PMID: 35145392 PMCID: PMC8821905 DOI: 10.3389/fnagi.2021.791679] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/27/2021] [Indexed: 11/13/2022] Open
Abstract
The microtubule-associated protein tau plays an important role in tauopathic diseases such as Alzheimer's disease and primary tauopathies such as progressive supranuclear palsy and corticobasal degeneration. Tauopathy animal models, such as transgenic, knock-in mouse and rat models, recapitulating tauopathy have facilitated the understanding of disease mechanisms. Aberrant accumulation of hyperphosphorylated tau contributes to synaptic deficits, neuroinflammation, and neurodegeneration, leading to cognitive impairment in animal models. Recent advances in molecular imaging using positron emission tomography (PET) and magnetic resonance imaging (MRI) have provided valuable insights into the time course of disease pathophysiology in tauopathy animal models. High-field MRI has been applied for in vivo imaging in animal models of tauopathy, including diffusion tensor imaging for white matter integrity, arterial spin labeling for cerebral blood flow, resting-state functional MRI for functional connectivity, volumetric MRI for neurodegeneration, and MR spectroscopy. In addition, MR contrast agents for non-invasive imaging of tau have been developed recently. Many preclinical MRI indicators offer excellent translational value and provide a blueprint for clinical MRI in the brains of patients with tauopathies. In this review, we summarized the recent advances in using MRI to visualize the pathophysiology of tauopathy in small animals. We discussed the outstanding challenges in brain imaging using MRI in small animals and propose a future outlook for visualizing tau-related alterations in the brains of animal models.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, ETH Zurich and University of Zurich, Zurich, Switzerland
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Cao L, Kong Y, Ji B, Ren Y, Guan Y, Ni R. Positron Emission Tomography in Animal Models of Tauopathies. Front Aging Neurosci 2022; 13:761913. [PMID: 35082657 PMCID: PMC8784812 DOI: 10.3389/fnagi.2021.761913] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/30/2021] [Indexed: 12/18/2022] Open
Abstract
The microtubule-associated protein tau (MAPT) plays an important role in Alzheimer's disease and primary tauopathy diseases. The abnormal accumulation of tau contributes to the development of neurotoxicity, inflammation, neurodegeneration, and cognitive deficits in tauopathy diseases. Tau synergically interacts with amyloid-beta in Alzheimer's disease leading to detrimental consequence. Thus, tau has been an important target for therapeutics development for Alzheimer's disease and primary tauopathy diseases. Tauopathy animal models recapitulating the tauopathy such as transgenic, knock-in mouse and rat models have been developed and greatly facilitated the understanding of disease mechanisms. The advance in PET and imaging tracers have enabled non-invasive detection of the accumulation and spread of tau, the associated microglia activation, metabolic, and neurotransmitter receptor alterations in disease animal models. In vivo microPET studies on mouse or rat models of tauopathy have provided significant insights into the phenotypes and time course of pathophysiology of these models and allowed the monitoring of treatment targeting at tau. In this study, we discuss the utilities of PET and recently developed tracers for evaluating the pathophysiology in tauopathy animal models. We point out the outstanding challenges and propose future outlook in visualizing tau-related pathophysiological changes in brain of tauopathy disease animal models.
Collapse
Affiliation(s)
- Lei Cao
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Changes Technology Corporation Ltd., Shanghai, China
| | - Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Bin Ji
- Department of Radiopharmacy and Molecular Imaging, School of Pharmacy, Fudan University, Shanghai, China
| | - Yutong Ren
- Guangdong Robotics Association, Guangzhou, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, ETH Zurich and University of Zurich, Zurich, Switzerland
| |
Collapse
|
14
|
Ni R, Nitsch RM. Recent Developments in Positron Emission Tomography Tracers for Proteinopathies Imaging in Dementia. Front Aging Neurosci 2022; 13:751897. [PMID: 35046791 PMCID: PMC8761855 DOI: 10.3389/fnagi.2021.751897] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022] Open
Abstract
An early detection and intervention for dementia represent tremendous unmet clinical needs and priorities in society. A shared feature of neurodegenerative diseases causing dementia is the abnormal accumulation and spreading of pathological protein aggregates, which affect the selective vulnerable circuit in a disease-specific pattern. The advancement in positron emission tomography (PET) biomarkers has accelerated the understanding of the disease mechanism and development of therapeutics for Alzheimer's disease and Parkinson's disease. The clinical utility of amyloid-β PET and the clinical validity of tau PET as diagnostic biomarker for Alzheimer's disease continuum have been demonstrated. The inclusion of biomarkers in the diagnostic criteria has introduced a paradigm shift that facilitated the early and differential disease diagnosis and impacted on the clinical management. Application of disease-modifying therapy likely requires screening of patients with molecular evidence of pathological accumulation and monitoring of treatment effect assisted with biomarkers. There is currently still a gap in specific 4-repeat tau imaging probes for 4-repeat tauopathies and α-synuclein imaging probes for Parkinson's disease and dementia with Lewy body. In this review, we focused on recent development in molecular imaging biomarkers for assisting the early diagnosis of proteinopathies (i.e., amyloid-β, tau, and α-synuclein) in dementia and discussed future perspectives.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, ETH & University of Zurich, Zurich, Switzerland
| | - Roger M. Nitsch
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
15
|
Filip T, Mairinger S, Neddens J, Sauberer M, Flunkert S, Stanek J, Wanek T, Okamura N, Langer O, Hutter-Paier B, Kuntner C. Characterization of an APP/tau rat model of Alzheimer's disease by positron emission tomography and immunofluorescent labeling. Alzheimers Res Ther 2021; 13:175. [PMID: 34656177 PMCID: PMC8522096 DOI: 10.1186/s13195-021-00916-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 10/05/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND To better understand the etiology and pathomechanisms of Alzheimer's disease, several transgenic animal models that overexpress human tau or human amyloid-beta (Aβ) have been developed. In the present study, we generated a novel transgenic rat model by cross-breeding amyloid precursor protein (APP) rats with tau rats. We characterized this model by performing positron emission tomography scans combined with immunofluorescent labeling and cerebrospinal fluid analyses. METHODS APP/Tau rats were generated by cross-breeding male McGill-R-Thy1-APP transgenic rats with female hTau-40/P301L transgenic rats. APP/Tau double transgenic rats and non-transgenic (ntg) littermates aged 7, 13, and 21 months were subjected to dynamic [11C] PiB scan and dynamic [18F]THK-5317 scans. For regional brain analysis, a template was generated from anatomical MR images of selected animals, which was co-registered with the PET images. Regional analysis was performed by application of the simplified reference tissue model ([11C]PiB data), whereas [18F]THK-5317 data were analyzed using a 2-tissue compartment model and Logan graphical analysis. In addition, immunofluorescent labeling (tau, amyloid) and cerebrospinal fluid analyses were performed. RESULTS [11C]PiB binding potential (BPND) and [18F]THK-5317 volume of distribution (VT) showed an increase with age in several brain regions in the APP/Tau group but not in the ntg control group. Immunohistochemical analysis of brain slices of PET-scanned animals revealed a positive correlation between Aβ labeling and [11C]PiB regional BPND. Tau staining yielded a trend towards higher levels in the cortex and hippocampus of APP/Tau rats compared with ntg littermates, but without reaching statistical significance. No correlation was found between tau immunofluorescence labeling results and the respective [18F]THK-5317 VT values. CONCLUSIONS We thoroughly characterized a novel APP/Tau rat model using combined PET imaging and immunofluorescence analysis. We observed an age-related increase in [11C]PiB and [18F]THK-5317 binding in several brain regions in the APP/Tau group but not in the ntg group. Although we were able to reveal a positive correlation between amyloid labeling and [11C]PiB regional brain uptake, we observed relatively low human tau and amyloid fibril expression levels and a somewhat unstable brain pathology which questions the utility of this animal model for further studies.
Collapse
Affiliation(s)
- Thomas Filip
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria
- Department of Biomedical Research, Medical University Vienna, Vienna, Austria
| | - Severin Mairinger
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Joerg Neddens
- Neuropharmacology, QPS Austria GmbH, Grambach, Austria
| | - Michael Sauberer
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | | | - Johann Stanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Thomas Wanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Oliver Langer
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | | | - Claudia Kuntner
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria.
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
16
|
Xiang X, Wind K, Wiedemann T, Blume T, Shi Y, Briel N, Beyer L, Biechele G, Eckenweber F, Zatcepin A, Lammich S, Ribicic S, Tahirovic S, Willem M, Deussing M, Palleis C, Rauchmann BS, Gildehaus FJ, Lindner S, Spitz C, Franzmeier N, Baumann K, Rominger A, Bartenstein P, Ziegler S, Drzezga A, Respondek G, Buerger K, Perneczky R, Levin J, Höglinger GU, Herms J, Haass C, Brendel M. Microglial activation states drive glucose uptake and FDG-PET alterations in neurodegenerative diseases. Sci Transl Med 2021; 13:eabe5640. [PMID: 34644146 DOI: 10.1126/scitranslmed.abe5640] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
2-Deoxy-2-[18F]fluoro-d-glucose positron emission tomography (FDG-PET) is widely used to study cerebral glucose metabolism. Here, we investigated whether the FDG-PET signal is directly influenced by microglial glucose uptake in mouse models and patients with neurodegenerative diseases. Using a recently developed approach for cell sorting after FDG injection, we found that, at cellular resolution, microglia displayed higher glucose uptake than neurons and astrocytes. Alterations in microglial glucose uptake were responsible for both the FDG-PET signal decrease in Trem2-deficient mice and the FDG-PET signal increase in mouse models for amyloidosis. Thus, opposite microglial activation states determine the differential FDG uptake. Consistently, 12 patients with Alzheimer’s disease and 21 patients with four-repeat tauopathies also exhibited a positive association between glucose uptake and microglial activity as determined by 18F-GE-180 18-kDa translocator protein PET (TSPO-PET) in preserved brain regions, indicating that the cerebral glucose uptake in humans is also strongly influenced by microglial activity. Our findings suggest that microglia activation states are responsible for FDG-PET signal alterations in patients with neurodegenerative diseases and mouse models for amyloidosis. Microglial activation states should therefore be considered when performing FDG-PET.
Collapse
Affiliation(s)
- Xianyuan Xiang
- Biomedical Center (BMC), Division of Metabolic Biochemistry, Faculty of Medicine, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, 518055 Shenzhen, China
| | - Karin Wind
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Thomas Wiedemann
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Tanja Blume
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Yuan Shi
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Nils Briel
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University München, 81377 Munich, Germany
| | - Leonie Beyer
- 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
| | - Florian Eckenweber
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Artem Zatcepin
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Sven Lammich
- Biomedical Center (BMC), Division of Metabolic Biochemistry, Faculty of Medicine, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Sara Ribicic
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Sabina Tahirovic
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Michael Willem
- Biomedical Center (BMC), Division of Metabolic Biochemistry, Faculty of Medicine, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Maximilian Deussing
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Carla Palleis
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Department of Neurology, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Boris-Stephan Rauchmann
- Department of Radiology, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Franz-Josef Gildehaus
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Charlotte Spitz
- Institute of Biochemistry and Molecular Biology, University of Augsburg, 86159 Augsburg, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Karlheinz Baumann
- Roche, Pharma Research and Early Development, NORD DTA/Neuroscience Discovery, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Axel Rominger
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- Department of Nuclear Medicine, University of Bern, Inselspital, CH-3010 Bern, Switzerland
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, 5091 Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE) Bonn-Cologne, 53127 Bonn, Germany
- Institute of Neuroscience and Medicine (INM-2), Molecular Organization of the Brain, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Gesine Respondek
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Katharina Buerger
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College, London SW7 2AZ, UK
| | - 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
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
- Department of Neurology, Technical University Munich, 81675 Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University München, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Christian Haass
- Biomedical Center (BMC), Division of Metabolic Biochemistry, Faculty of Medicine, Ludwig-Maximilians-Universität München, 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
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| |
Collapse
|
17
|
(S)-[ 18F]THK5117 brain uptake is associated with Aβ plaques and MAO-B enzyme in a mouse model of Alzheimer's disease. Neuropharmacology 2021; 196:108676. [PMID: 34216585 DOI: 10.1016/j.neuropharm.2021.108676] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 12/12/2022]
Abstract
The mouse model of beta-amyloid (Aβ) deposition, APP/PS1-21, exhibits high brain uptake of the tau-tracer (S)-[18F]THK5117, although no neurofibrillary tangles are present in this mouse model. For this reason we investigated (S)-[18F]THK5117 off-target binding to Aβ plaques and MAO-B enzyme in APP/PS1-21 transgenic (TG) mouse model of Aβ deposition. APP/PS1-21 TG and wild-type (WT) control mice in four different age groups (2-26 months) were imaged antemortem by positron emission tomography with (S)-[18F]THK5117, and then brain autoradiography. Additional animals were used for immunohistochemical staining and MAO-B enzyme blocking study with deprenyl pre-treatment. Regional standardized uptake value ratios for the cerebellum revealed a significant temporal increase in (S)-[18F]THK5117 uptake in aged TG, but not WT, brain. Immunohistochemical staining revealed a similar increase in Aβ plaques but not endogenous hyper-phosphorylated tau or MAO-B enzyme, and ex vivo autography showed that uptake of (S)-[18F]THK5117 co-localized with the amyloid pathology. Deprenyl hydrochloride pre-treatment reduced the binding of (S)-[18F]THK5117 in the neocortex, hippocampus, and thalamus. This study's findings suggest that increased (S)-[18F]THK5117 binding in aging APP/PS1-21 TG mice is mainly due to increasing Aβ deposition, and to a lesser extent binding to MAO-B enzyme, but not hyper-phosphorylated tau.
Collapse
|
18
|
McMurray L, Macdonald JA, Ramakrishnan NK, Zhao Y, Williamson DW, Tietz O, Zhou X, Kealey S, Fagan SG, Smolek T, Cubinkova V, Žilka N, Spillantini MG, Tolkovsky AM, Goedert M, Aigbirhio FI. Synthesis and Assessment of Novel Probes for Imaging Tau Pathology in Transgenic Mouse and Rat Models. ACS Chem Neurosci 2021; 12:1885-1893. [PMID: 33689290 PMCID: PMC8176454 DOI: 10.1021/acschemneuro.0c00790] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Aggregated tau protein is a core pathology present in several neurodegenerative diseases. Therefore, the development and application of positron emission tomography (PET) imaging radiotracers that selectively bind to aggregated tau in fibril form is of importance in furthering the understanding of these disorders. While radiotracers used in human PET studies offer invaluable insight, radiotracers that are also capable of visualizing tau fibrils in animal models are important tools for translational research into these diseases. Herein, we report the synthesis and characterization of a novel library of compounds based on the phenyl/pyridinylbutadienylbenzothiazoles/benzothiazolium (PBB3) backbone developed for this application. From this library, we selected the compound LM229, which binds to recombinant tau fibrils with high affinity (Kd = 3.6 nM) and detects with high specificity (a) pathological 4R tau aggregates in living cultured neurons and mouse brain sections from transgenic human P301S tau mice, (b) truncated human 151-351 3R (SHR24) and 4R (SHR72) tau aggregates in transgenic rat brain sections, and (c) tau neurofibrillary tangles in brain sections from Alzheimer's disease (3R/4R tau) and progressive supranuclear palsy (4R tau). With LM229 also shown to cross the blood-brain barrier in vivo and its effective radiolabeling with the radioisotope carbon-11, we have established a novel platform for PET translational studies using rodent transgenic tau models.
Collapse
Affiliation(s)
- Lindsay McMurray
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | | | - Nisha Kuzhuppilly Ramakrishnan
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Yanyan Zhao
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - David W. Williamson
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Ole Tietz
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Xiaoyun Zhou
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Steven Kealey
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Steven G. Fagan
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Tomáš Smolek
- Axon Neuroscience R&D Services SE, Bratislava, Slovak Republic 811 02
| | | | - Norbert Žilka
- Axon Neuroscience R&D Services SE, Bratislava, Slovak Republic 811 02
| | - Maria Grazia Spillantini
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Aviva M. Tolkovsky
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Michel Goedert
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Franklin I. Aigbirhio
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
19
|
Bertoglio D, Verhaeghe J, Miranda A, Wyffels L, Stroobants S, Dominguez C, Munoz-Sanjuan I, Skinbjerg M, Liu L, Staelens S. Kinetic Modelling and Test-Retest Reproducibility for the Dopamine D 1R Radioligand [ 11C]SCH23390 in Healthy and Diseased Mice. Mol Imaging Biol 2021; 23:208-219. [PMID: 33179158 PMCID: PMC7910372 DOI: 10.1007/s11307-020-01561-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/29/2020] [Accepted: 11/03/2020] [Indexed: 01/25/2023]
Abstract
PURPOSE Our aim in this study was to compare different non-invasive pharmacokinetic models and assess test-retest reproducibility of the radioligand [11C]SCH23390 for the quantification of dopamine D1-like receptor (D1R) in both wild-type (WT) mice and heterozygous (HET) Q175DN mice as Huntington's disease (HD) model. PROCEDURES Adult WT (n = 9) and HET (n = 14) mice underwent a 90-min [11C]SCH23390 positron emission tomography (PET) scan followed by computed tomography (CT) to evaluate the pharmacokinetic modelling in healthy and diseased conditions. Additionally, 5 WT mice and 7 HET animals received a second [11C]SCH23390 PET scan for test-retest reproducibility. Parallel assessment of the simplified reference tissue model (SRTM), the multilinear reference tissue model (MRTM) and the Logan reference tissue model (Logan Ref) using the striatum as a receptor-rich region and the cerebellum as a receptor-free (reference) region was performed to define the most suitable method for regional- and voxel-based quantification of the binding potential (BPND). Finally, standardised uptake value ratio (SUVR-1) was assessed as a potential simplified measurement. RESULTS For all models, we measured a significant decline in dopamine D1R density (e.g. SRTM = - 38.5 ± 5.0 %, p < 0.0001) in HET mice compared to WT littermates. Shortening the 90-min scan duration resulted in large underestimation of striatal BPND in both WT mice (SRTM 60 min: - 17.7 ± 2.8 %, p = 0.0078) and diseased HET (SRTM 60 min: - 13.1 ± 4.1 %, p = 0.0001). Striatal BPND measurements were very reproducible with an average test-retest variability below 5 % when using both MRTM and SRTM. Parametric BPND maps generated with SRTM were highly reliable, showing nearly perfect agreement to the regional analysis (r2 = 0.99, p < 0.0001). Finally, SRTM provided the most accurate estimate for relative tracer delivery R1 with both regional- and voxel-based analyses. SUVR-1 at different time intervals were not sufficiently reliable when compared to BPND (r2 < 0.66). CONCLUSIONS Ninety-minute acquisition and the use of SRTM for pharmacokinetic modelling is recommended. [11C]SCH23390 PET imaging demonstrates optimal characteristics for the study of dopamine D1R density in models of psychiatric and neurological disorders as exemplified in the Q175DN mouse model of HD.
Collapse
Affiliation(s)
- Daniele Bertoglio
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium
| | - Jeroen Verhaeghe
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium
| | - Alan Miranda
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium
| | - Leonie Wyffels
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium
- Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium
- Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | | | | | | | - Longbin Liu
- CHDI Management/CHDI Foundation, Los Angeles, CA, USA
| | - Steven Staelens
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium.
| |
Collapse
|
20
|
Abstract
Positron emission tomography (PET) is a non-invasive imaging technology employed to describe metabolic, physiological, and biochemical processes in vivo. These include receptor availability, metabolic changes, neurotransmitter release, and alterations of gene expression in the brain. Since the introduction of dedicated small-animal PET systems along with the development of many novel PET imaging probes, the number of PET studies using rats and mice in basic biomedical research tremendously increased over the last decade. This article reviews challenges and advances of quantitative rodent brain imaging to make the readers aware of its physical limitations, as well as to inspire them for its potential applications in preclinical research. In the first section, we briefly discuss the limitations of small-animal PET systems in terms of spatial resolution and sensitivity and point to possible improvements in detector development. In addition, different acquisition and post-processing methods used in rodent PET studies are summarized. We further discuss factors influencing the test-retest variability in small-animal PET studies, e.g., different receptor quantification methodologies which have been mainly translated from human to rodent receptor studies to determine the binding potential and changes of receptor availability and radioligand affinity. We further review different kinetic modeling approaches to obtain quantitative binding data in rodents and PET studies focusing on the quantification of endogenous neurotransmitter release using pharmacological interventions. While several studies have focused on the dopamine system due to the availability of several PET tracers which are sensitive to dopamine release, other neurotransmitter systems have become more and more into focus and are described in this review, as well. We further provide an overview of latest genome engineering technologies, including the CRISPR/Cas9 and DREADD systems that may advance our understanding of brain disorders and function and how imaging has been successfully applied to animal models of human brain disorders. Finally, we review the strengths and opportunities of simultaneous PET/magnetic resonance imaging systems to study drug-receptor interactions and challenges for the translation of PET results from bench to bedside.
Collapse
|
21
|
Kelberman M, Keilholz S, Weinshenker D. What's That (Blue) Spot on my MRI? Multimodal Neuroimaging of the Locus Coeruleus in Neurodegenerative Disease. Front Neurosci 2020; 14:583421. [PMID: 33122996 PMCID: PMC7573566 DOI: 10.3389/fnins.2020.583421] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/16/2020] [Indexed: 01/04/2023] Open
Abstract
The locus coeruleus (LC) has long been underappreciated for its role in the pathophysiology of Alzheimer’s disease (AD), Parkinson’s disease (PD), and other neurodegenerative disorders. While AD and PD are distinct in clinical presentation, both are characterized by prodromal protein aggregation in the LC, late-stage degeneration of the LC, and comorbid conditions indicative of LC dysfunction. Many of these early studies were limited to post-mortem histological techniques due to the LC’s small size and location deep in the brainstem. Thus, there is a growing interest in utilizing in vivo imaging of the LC as a predictor of preclinical neurodegenerative processes and biomarker of disease progression. Simultaneously, neuroimaging in animal models of neurodegenerative disease holds promise for identifying early alterations to LC circuits, but has thus far been underutilized. While still in its infancy, a handful of studies have reported effects of single gene mutations and pathology on LC function in disease using various neuroimaging techniques. Furthermore, combining imaging and optogenetics or chemogenetics allows for interrogation of network connectivity in response to changes in LC activity. The purpose of this article is twofold: (1) to review what magnetic resonance imaging (MRI) and positron emission tomography (PET) have revealed about LC dysfunction in neurodegenerative disease and its potential as a biomarker in humans, and (2) to explore how animal models can be used to test hypotheses derived from clinical data and establish a mechanistic framework to inform LC-focused therapeutic interventions to alleviate symptoms and impede disease progression.
Collapse
Affiliation(s)
- Michael Kelberman
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| | - Shella Keilholz
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - David Weinshenker
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| |
Collapse
|
22
|
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
|
23
|
Arora H, Ramesh M, Rajasekhar K, Govindaraju T. Molecular Tools to Detect Alloforms of Aβ and Tau: Implications for Multiplexing and Multimodal Diagnosis of Alzheimer’s Disease. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2020. [DOI: 10.1246/bcsj.20190356] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Harshit Arora
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru 560064, Karnataka, India
| | - Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru 560064, Karnataka, India
| | - Kolla Rajasekhar
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru 560064, Karnataka, India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru 560064, Karnataka, India
- VNIR Biotechnologies Pvt. Ltd., Bangalore Bioinnovation Center, Helix Biotech Park, Electronic City Phase I, Bengaluru 560100, Karnataka, India
| |
Collapse
|
24
|
Weng CC, Hsiao IT, Yang QF, Yao CH, Tai CY, Wu MF, Yen TC, Jang MK, Lin KJ. Characterization of 18F-PM-PBB3 ( 18F-APN-1607) Uptake in the rTg4510 Mouse Model of Tauopathy. Molecules 2020; 25:molecules25071750. [PMID: 32290239 PMCID: PMC7181044 DOI: 10.3390/molecules25071750] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 12/03/2022] Open
Abstract
Misfolding, aggregation, and cerebral accumulation of tau deposits are hallmark features of Alzheimer’s disease. Positron emission tomography study of tau can facilitate the development of anti-tau treatment. Here, we investigated a novel tau tracer 18F-PM-PBB3 (18F-APN-1607) in a mouse model of tauopathy. Dynamic PET scans were collected in groups of rTg4510 transgenic mice at 2–11 months of age. Associations between distribution volume ratios (DVR) and standardized uptake value ratios (SUVR) with cerebellum reference were used to determine the optimal scanning time and uptake pattern for each age. Immunohistochemistry staining of neurofibrillary tangles and autoradiography study was performed for ex vivo validation. An SUVR 40–70 min was most consistently correlated with DVR and was used in further analyses. Significant increased 18F-PM-PBB3 uptake in the brain cortex was found in six-month-old mice (+28.9%, p < 0.05), and increased further in the nine-month-old group (+38.8%, p < 0.01). The trend of increased SUVR value remained evident in the hippocampus and striatum regions except for cortex where uptake becomes slightly reduced in 11-month-old animals (+37.3%, p < 0.05). Radioactivity distributions from autoradiography correlate well to the presence of human tau (HT7 antibody) and hyperphosphorylated tau (antibody AT8) from the immunohistochemistry study of the adjacent brain sections. These findings supported that the 40–70 min 18F-PM-PBB3 PET scan with SUVR measurement can detect significantly increased tau deposits in a living rTg4510 transgenic mouse models as early as six-months-old. The result exhibited promising dynamic imaging capability of this novel tau tracer, and the above image characteristics should be considered in the design of longitudinal preclinical tau image studies.
Collapse
Affiliation(s)
- Chi-Chang Weng
- HARC and Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan 333, Taiwan; (C.C.-W.); (I.-T.H.); (Q.-F.Y.)
- Department of Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Ing-Tsung Hsiao
- HARC and Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan 333, Taiwan; (C.C.-W.); (I.-T.H.); (Q.-F.Y.)
- Department of Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Qing-Fang Yang
- HARC and Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan 333, Taiwan; (C.C.-W.); (I.-T.H.); (Q.-F.Y.)
| | - Cheng-Hsiang Yao
- Department of Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Chin-Yin Tai
- APRINOIA Therapeutics Inc., Taipei 11503, Taiwan; (C.-Y.T.); (M.-F.W.); (T.-C.Y.); (M.-K.J.)
| | - Meng-Fang Wu
- APRINOIA Therapeutics Inc., Taipei 11503, Taiwan; (C.-Y.T.); (M.-F.W.); (T.-C.Y.); (M.-K.J.)
| | - Tzu-Chen Yen
- APRINOIA Therapeutics Inc., Taipei 11503, Taiwan; (C.-Y.T.); (M.-F.W.); (T.-C.Y.); (M.-K.J.)
| | - Ming-Kuei Jang
- APRINOIA Therapeutics Inc., Taipei 11503, Taiwan; (C.-Y.T.); (M.-F.W.); (T.-C.Y.); (M.-K.J.)
| | - Kun-Ju Lin
- Department of Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- Correspondence:
| |
Collapse
|
25
|
Edwards G, Zhao J, Dash PK, Soto C, Moreno-Gonzalez I. Traumatic Brain Injury Induces Tau Aggregation and Spreading. J Neurotrauma 2019; 37:80-92. [PMID: 31317824 PMCID: PMC6921297 DOI: 10.1089/neu.2018.6348] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The misfolding and aggregation of tau protein into neurofibrillary tangles is the main underlying hallmark of tauopathies. Most tauopathies have a sporadic origin and can be associated with multiple risk factors. Traumatic brain injury (TBI) has been suggested as a risk factor for tauopathies by triggering disease onset and facilitating its progression. Several studies indicate that TBI seems to be a risk factor to development of Alzheimer disease and chronic traumatic encephalopathy, because there is a relationship of TBI severity and propensity to development of these illnesses. In this study, we evaluated whether moderate to severe TBI can trigger the initial formation of pathological tau that would induce the development of the pathology throughout the brain. To this end, we subjected tau transgenic mice to TBI and assessed tau phosphorylation and aggregation pattern to create a spatial heat map of tau deposition and spreading in the brain. Our results suggest that brain injured tau transgenic mice have an accelerated tau pathology in different brain regions that increases over time compared with sham mice. The appearance of pathological tau occurs in regions distant to the injury area that are connected synaptically, suggesting dissemination of tau aggregates. Overall, this work posits TBI as a risk factor for tauopathies through the induction of tau hyperphosphorylation and aggregation.
Collapse
Affiliation(s)
- George Edwards
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Jing Zhao
- Department of Neurobiology and Anatomy, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Pramod K Dash
- Department of Neurobiology and Anatomy, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Claudio Soto
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Ines Moreno-Gonzalez
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center at Houston, Houston, Texas.,Department of Cell Biology, Networking Research Center on Neurodegenerative Diseases (CIBERNED), Facultad Ciencias, Universidad de Malaga, Malaga, Spain
| |
Collapse
|
26
|
Lee M, Lee HJ, Jeong YJ, Oh SJ, Kang KJ, Han SJ, Nam KR, Lee YJ, Lee KC, Ryu YH, Hyun IY, Choi JY. Age dependency of mGluR5 availability in 5xFAD mice measured by PET. Neurobiol Aging 2019; 84:208-216. [PMID: 31570178 DOI: 10.1016/j.neurobiolaging.2019.08.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 07/29/2019] [Accepted: 08/06/2019] [Indexed: 11/25/2022]
Abstract
The major pathologies of Alzheimer's disease (AD) are amyloid plaques and hyperphosphorylated tau. The deposition of amyloid plaques leads to synaptic dysfunction, neuronal cell death, and cognitive impairment. Among the neurotransmitters, glutamate is the most abundant in the mammalian brain and plays an important role in synaptic plasticity. With respect to synaptic transmission, metabotropic glutamate receptor 5 (mGluR5) is highly affected by amyloid pathology. However, the neuropathologic changes in the protein expression of mGluR5 in AD remain unclear. Therefore, to elucidate the alteration in mGluR5 expression with the progression of AD, we performed serial behavioral tests, longitudinal imaging studies, and histopathological immunoassay for both 5xFAD (n = 14) mice and age-matched wild-type mice (n = 14). The 5xFAD mice started showing severe hyperactivity and memory impairment from 7 months of age. In addition, mGluR5 positron emission tomography revealed that while the binding values in the wild-type mice were similar over time, those in 5xFAD mice fluctuated from 5 months of age. Furthermore, the 5xFAD mice presented a 35% decrease in the binding values of their cortical and subcortical areas at 9 months of age compared with those at 3 months of age. Magnetic resonance spectroscopy and histopathological studies showed similar changes. In conclusion, mGluR5 availability changes with age, and mGluR5 positron emission tomography could successfully detect this synaptic change in the 5xFAD mice.
Collapse
Affiliation(s)
- Minkyung Lee
- Department of Nuclear Medicine, Inha University Hospital, Inha University, Incheon, South Korea
| | - Hae-June Lee
- Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Ye Ji Jeong
- Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Se Jong Oh
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Kyung Jun Kang
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Sang Jin Han
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Kyung Rok Nam
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Yong Jin Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Kyo Chul Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Young Hoon Ryu
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - In Young Hyun
- Department of Nuclear Medicine, Inha University Hospital, Inha University, Incheon, South Korea
| | - Jae Yong Choi
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea.
| |
Collapse
|
27
|
Brendel M, Deussing M, Blume T, Kaiser L, Probst F, Overhoff F, Peters F, von Ungern-Sternberg B, Ryazanov S, Leonov A, Griesinger C, Zwergal A, Levin J, Bartenstein P, Yakushev I, Cumming P, Boening G, Ziegler S, Herms J, Giese A, Rominger A. Late-stage Anle138b treatment ameliorates tau pathology and metabolic decline in a mouse model of human Alzheimer's disease tau. Alzheimers Res Ther 2019; 11:67. [PMID: 31370885 PMCID: PMC6670231 DOI: 10.1186/s13195-019-0522-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 07/22/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND Augmenting the brain clearance of toxic oligomers with small molecule modulators constitutes a promising therapeutic concept against tau deposition. However, there has been no test of this concept in animal models of Alzheimer's disease (AD) with initiation at a late disease stage. Thus, we aimed to investigate the effects of interventional late-stage Anle138b treatment, which previously indicated great potential to inhibit oligomer accumulation by binding of pathological aggregates, on the metabolic decline in transgenic mice with established tauopathy in a longitudinal 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) study. METHODS Twelve transgenic mice expressing all six human tau isoforms (hTau) and ten controls were imaged by FDG-PET at baseline (14.5 months), followed by randomization into Anle138b treatment and vehicle groups for 3 months. FDG-PET was repeated after treatment for 3 months, and brains were analyzed by tau immunohistochemistry. Longitudinal changes of glucose metabolism were compared between study groups, and the end point tau load was correlated with individual FDG-PET findings. RESULTS Tau pathology was significantly ameliorated by late-stage Anle138b treatment when compared to vehicle (frontal cortex - 53%, p < 0.001; hippocampus - 59%, p < 0.005). FDG-PET revealed a reversal of metabolic decline during Anle138b treatment, whereas the vehicle group showed ongoing deterioration. End point glucose metabolism in the brain of hTau mice had a strong correlation with tau deposition measured by immunohistochemistry (R = 0.92, p < 0.001). CONCLUSION Late-stage oligomer modulation effectively ameliorated tau pathology in hTau mice and rescued metabolic function. Molecular imaging by FDG-PET can serve for monitoring effects of Anle138b treatment.
Collapse
Affiliation(s)
- Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr.15, 81377 Munich, Germany
| | - Maximilian Deussing
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr.15, 81377 Munich, Germany
| | - Tanja Blume
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr.15, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Lena Kaiser
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr.15, 81377 Munich, Germany
| | - Federico Probst
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr.15, 81377 Munich, Germany
| | - Felix Overhoff
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr.15, 81377 Munich, Germany
| | - Finn Peters
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | | | - Sergey Ryazanov
- Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Andrei Leonov
- Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
- MODAG GmbH, 55324 Wendelsheim, Germany
| | - Christian Griesinger
- Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
- DFG Research Centre Nanoscale Microscopy and Molecular Physiology of the Brain, 37070 Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Andreas Zwergal
- Department of Neurology, University Hospital, LMU Munich, Munich, Germany
| | - Johannes Levin
- Department of Neurology, University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr.15, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Igor Yakushev
- Neuroimaging Center (TUM-NIC), Technische Universität München, Munich, Germany
- Department of Nuclear Medicine, Technical University of Munich, 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
| | - Guido Boening
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr.15, 81377 Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr.15, 81377 Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-Universität, Feodor Lynen-Str. 23, 81377 Munich, Germany
| | - Armin Giese
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- MODAG GmbH, 55324 Wendelsheim, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr.15, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Nuclear Medicine, Inselspital Bern, Bern, Switzerland
| |
Collapse
|
28
|
Rösler TW, Tayaranian Marvian A, Brendel M, Nykänen NP, Höllerhage M, Schwarz SC, Hopfner F, Koeglsperger T, Respondek G, Schweyer K, Levin J, Villemagne VL, Barthel H, Sabri O, Müller U, Meissner WG, Kovacs GG, Höglinger GU. Four-repeat tauopathies. Prog Neurobiol 2019; 180:101644. [PMID: 31238088 DOI: 10.1016/j.pneurobio.2019.101644] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/21/2019] [Accepted: 06/12/2019] [Indexed: 02/08/2023]
Abstract
Tau is a microtubule-associated protein with versatile functions in the dynamic assembly of the neuronal cytoskeleton. Four-repeat (4R-) tauopathies are a group of neurodegenerative diseases defined by cytoplasmic inclusions predominantly composed of tau protein isoforms with four microtubule-binding domains. Progressive supranuclear palsy, corticobasal degeneration, argyrophilic grain disease or glial globular tauopathy belong to the group of 4R-tauopathies. The present review provides an introduction in the current concept of 4R-tauopathies, including an overview of the neuropathological and clinical spectrum of these diseases. It describes the genetic and environmental etiological factors, as well as the contemporary knowledge about the pathophysiological mechanisms, including post-translational modifications, aggregation and fragmentation of tau, as well as the role of protein degradation mechanisms. Furthermore, current theories about disease propagation are discussed, involving different extracellular tau species and their cellular release and uptake mechanisms. Finally, molecular diagnostic tools for 4R-tauopathies, including tau-PET and fluid biomarkers, and investigational therapeutic strategies are presented. In summary, we report on 4R-tauopathies as overarching disease concept based on a shared pathophysiological concept, and highlight the challenges and opportunities on the way towards a causal therapy.
Collapse
Affiliation(s)
- Thomas W Rösler
- Dept. of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Dept. of Neurology, Technical University of Munich, School of Medicine, 81675 Munich, Germany
| | - Amir Tayaranian Marvian
- Dept. of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Dept. of Neurology, Technical University of Munich, School of Medicine, 81675 Munich, Germany
| | - Matthias Brendel
- Dept. of Nuclear Medicine, University of Munich, 81377 Munich, Germany
| | - Niko-Petteri Nykänen
- Dept. of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Matthias Höllerhage
- Dept. of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Dept. of Neurology, Technical University of Munich, School of Medicine, 81675 Munich, Germany
| | - Sigrid C Schwarz
- Dept. of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | | | - Thomas Koeglsperger
- Dept. of Neurology, University of Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Gesine Respondek
- Dept. of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Dept. of Neurology, Technical University of Munich, School of Medicine, 81675 Munich, Germany
| | - Kerstin Schweyer
- Dept. of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Dept. of Neurology, Technical University of Munich, School of Medicine, 81675 Munich, Germany
| | - Johannes Levin
- Dept. of Neurology, University of Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Victor L Villemagne
- Dept. of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, 3084, Australia; The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia; Dept. of Medicine, Austin Health, University of Melbourne, Melbourne, VIC, Australia
| | - Henryk Barthel
- Dept. of Nuclear Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Osama Sabri
- Dept. of Nuclear Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Ulrich Müller
- Institute for Human Genetics, University of Giessen, 35392 Giessen, Germany
| | - Wassilios G Meissner
- Service de Neurologie, CHU Bordeaux, 33000 Bordeaux, France; Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Dept. of Medicine, University of Otago, Christchurch, New Zealand; New Zealand Brain Research Institute, Christchurch, New Zealand
| | - Gabor G Kovacs
- Institute of Neurology, Medical University of Vienna, 1090 Vienna, Austria; Dept. of Laboratory Medicine and Pathobiology, University of Toronto, Laboratory Medicine Program, University Health Network, Toronto, Canada; Tanz Centre for Research in Neurodegenerative Disease, Krembil Brain Institute, Toronto, Canada
| | - Günter U Höglinger
- Dept. of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Dept. of Neurology, Technical University of Munich, School of Medicine, 81675 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany; Dept. of Neurology, Hannover Medical School, 30625 Hannover, Germany.
| |
Collapse
|
29
|
Brendel M, Yousefi BH, Blume T, Herz M, Focke C, Deussing M, Peters F, Lindner S, von Ungern-Sternberg B, Drzezga A, Bartenstein P, Haass C, Okamura N, Herms J, Yakushev I, Rominger A. Comparison of 18F-T807 and 18F-THK5117 PET in a Mouse Model of Tau Pathology. Front Aging Neurosci 2018; 10:174. [PMID: 29930508 PMCID: PMC5999706 DOI: 10.3389/fnagi.2018.00174] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 05/22/2018] [Indexed: 11/16/2022] Open
Abstract
Positron-emission-tomography (PET) imaging of tau pathology has facilitated development of anti-tau therapies. While members of the arylquinoline and pyridoindole families have been the most frequently used tau radioligands so far, analyses of their comparative performance in vivo are scantly documented. Here, we conducted a head-to-head PET comparison of the arylquinoline 18FT807 and the pyridoindole 18FTHK5117 PET in a mouse model of tau pathology. PET recordings were obtained in groups of (N = 5–7) P301S and wild-type (WT) mice at 6 and 9 months of age. Volume-of-interest based analysis (standard-uptake-value ratio, SUVR) was used to calculate effect sizes (Cohen’s d) for each tracer and age. Statistical parametric mapping (SPM) was used to assess regional similarity (dice coefficient) of tracer binding alterations for the two tracers. Immunohistochemistry staining of neurofibrillary tangles was performed for validation ex vivo. Significantly elevated 18F-T807 binding in the brainstem of P301S mice was already evident at 6 months (+14%, p < 0.01, d = 1.64), and increased further at 9 months (+23%, p < 0.001, d = 2.70). 18F-THK5117 indicated weaker increases and effect sizes at 6 months (+5%, p < 0.05, d = 1.07) and 9 months (+10%, p < 0.001, d = 1.49). Regional similarity of binding of the two tracers was high (71%) at 9 months. 18F-T807 was more sensitive than 18F-THK5117 to tau pathology in this model, although both tracers present certain obstacles, which need to be considered in the design of longitudinal preclinical tau imaging studies.
Collapse
Affiliation(s)
- Matthias Brendel
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Behrooz H Yousefi
- Department of Nuclear Medicine, Technical University of Munich, Munich, Germany.,Neuroimaging Center, Technische Universität München, Munich, Germany
| | - Tanja Blume
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Michael Herz
- Department of Nuclear Medicine, Technical University of Munich, Munich, Germany
| | - Carola Focke
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Maximilian Deussing
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Finn Peters
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | | | - Alexander Drzezga
- Department of Nuclear Medicine, University of Cologne, Cologne, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Biomedical Center, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Igor Yakushev
- Department of Nuclear Medicine, Technical University of Munich, Munich, Germany.,Neuroimaging Center, Technische Universität München, Munich, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
30
|
Ni R, Ji B, Ono M, Sahara N, Zhang MR, Aoki I, Nordberg A, Suhara T, Higuchi M. Comparative In Vitro and In Vivo Quantifications of Pathologic Tau Deposits and Their Association with Neurodegeneration in Tauopathy Mouse Models. J Nucl Med 2018; 59:960-966. [PMID: 29419480 DOI: 10.2967/jnumed.117.201632] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 01/09/2018] [Indexed: 11/16/2022] Open
Abstract
Fibrillary tau aggregates in Alzheimer disease and allied neurodegenerative disorders have been visualized in vivo by PET, whereas mechanistic links between PET-detectable tau deposits and neurotoxicity remain elusive. Here, we took advantage of transgenic mouse models of tauopathies to evaluate associations between PET and postmortem measures of tau probe binding and their relation to neuronal loss. Methods: PET with a tau probe, 11C-PBB3 (2-((1E,3E)-4-(6-(11C-methylamino)pyridine-3-yl)buta-1,3-dienyl)benzo[d]thiazol-6-ol), and volumetric MRI were performed for transgenic rTg4510 mice and nontransgenic mice. Binding of 11C-PBB3 and its blockade by another tau binding compound, AV-1451 (-(6-fluoropyridine-3-yl)-5H-pyrido[4,3-b]indole), in homogenized brains of tauopathy patients and rTg4510 and PS19 mice were quantified, and 11C-PBB3-positive and phosphorylated tau lesions in sectioned brains of these mice were assessed. Results: In vivo 11C-PBB3 binding to the rTg4510 neocortex/hippocampus was increased relative to controls and correlated with local atrophy. In vitro 11C-PBB3 binding in the neocortex/hippocampus also correlated well with in vivo radioligand binding and regional atrophy in the same individual rTg4510 mice. By contrast, in vitro 11C-PBB3 binding was elevated in the brain stem but not hippocampus of PS19 mice, despite a pronounced loss of neurons in the hippocampus rather than brain stem. Finally, 11C-PBB3 and AV-1451 showed similar binding properties between mouse models and tauopathy patients. Conclusion: The present findings support the distinct utilities of 11C-PBB3 PET and MRI in rTg4510 and PS19 mice for quantitatively pursuing mechanisms connecting PET-detectable and PET-undetectable tau aggregations to neuronal death, which recapitulate 2 different modes of tau-provoked neurotoxicity.
Collapse
Affiliation(s)
- Ruiqing Ni
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan.,Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Karolinska Institute, Stockholm, Sweden; and.,Theme Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Bin Ji
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
| | - Maiko Ono
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
| | - Naruhiko Sahara
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
| | - Ming-Rong Zhang
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
| | - Ichio Aoki
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
| | - Agneta Nordberg
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Karolinska Institute, Stockholm, Sweden; and.,Theme Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Tetsuya Suhara
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
| | - Makoto Higuchi
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
| |
Collapse
|
31
|
Rajasekhar K, Govindaraju T. Current progress, challenges and future prospects of diagnostic and therapeutic interventions in Alzheimer's disease. RSC Adv 2018; 8:23780-23804. [PMID: 35540246 PMCID: PMC9081849 DOI: 10.1039/c8ra03620a] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 06/04/2018] [Indexed: 01/04/2023] Open
Abstract
The diverse pathological mechanisms and their implications for the development of effective diagnostic and therapeutic interventions in Alzheimer's disease are presented with current progress, challenges and future prospects.
Collapse
Affiliation(s)
- K. Rajasekhar
- Bioorganic Chemistry Laboratory
- New Chemistry Unit
- Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR)
- Bengaluru 560064
- India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory
- New Chemistry Unit
- Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR)
- Bengaluru 560064
- India
| |
Collapse
|
32
|
Sahara N, Shimojo M, Ono M, Takuwa H, Febo M, Higuchi M, Suhara T. In Vivo Tau Imaging for a Diagnostic Platform of Tauopathy Using the rTg4510 Mouse Line. Front Neurol 2017; 8:663. [PMID: 29375461 PMCID: PMC5770623 DOI: 10.3389/fneur.2017.00663] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 11/23/2017] [Indexed: 12/23/2022] Open
Abstract
Association of tau deposition with neurodegeneration in Alzheimer's disease (AD) and related tau-positive neurological disorders collectively referred to as tauopathies indicates contribution of tau aggregates to neurotoxicity. The discovery of tau gene mutations in FTDP-17-tau kindreds has provided unequivocal evidence that tau abnormalities alone can induce neurodegenerative disorders. Therefore, visualization of tau accumulation would offer a reliable, objective index to aid in the diagnosis of tauopathy and to assess the disease progression. Positron emission tomography (PET) imaging of tau lesions is currently available using several tau PET ligands. Because most tau PET ligands have the property of an extrinsic fluorescent dye, these ligands are considered to be useful for both PET and fluorescence imaging. In addition, small-animal magnetic resonance imaging (MRI) is available for both structural and functional imaging. Using these advanced imaging techniques, in vivo studies on a mouse model of tauopathy will provide significant insight into the translational research of neurodegenerative diseases. In this review, we will discuss the utilities of PET, MRI, and fluorescence imaging for evaluating the disease progression of tauopathy.
Collapse
Affiliation(s)
- Naruhiko Sahara
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Masafumi Shimojo
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Maiko Ono
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Hiroyuki Takuwa
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Marcelo Febo
- Department of Psychiatry and Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States
| | - Makoto Higuchi
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Tetsuya Suhara
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
33
|
Bischof GN, Endepols H, van Eimeren T, Drzezga A. Tau-imaging in neurodegeneration. Methods 2017; 130:114-123. [PMID: 28790016 DOI: 10.1016/j.ymeth.2017.08.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/25/2017] [Accepted: 08/04/2017] [Indexed: 10/19/2022] Open
Abstract
Pathological cerebral aggregations of proteins are suggested to play a crucial role in the development of neurodegenerative disorders. For example, aggregation of the protein ß-amyloid in form of extracellular amyloid-plaques as well as intraneuronal depositions of the protein tau in form of neurofibrillary tangles represent hallmarks of Alzheimer's disease (AD). Recently, novel tracers for in vivo molecular imaging of tau-aggregates in the brain have been introduced, complementing existing tracers for imaging amyloid-plaques. Available data on these novel tracers indicate that the subject of Tau-PET may be of considerable complexity. On the one hand this refers to the various forms of appearance of tau-pathology in different types of neurodegenerative disorders. On the other hand, a number of hurdles regarding validation of these tracers still need to be overcome with regard to comparability and standardization of the different tracers, observed off-target/non-specific binding and quantitative interpretation of the signal. These issues will have to be clarified before systematic clinical application of this exciting new methodological approach may become possible. Potential applications refer to early detection of neurodegeneration, differential diagnosis between tauopathies and non-tauopathies and specific patient selection and follow-up in therapy trials.
Collapse
Affiliation(s)
| | - Heike Endepols
- Department of Nuclear Medicine, University of Cologne, Germany
| | - Thilo van Eimeren
- Department of Nuclear Medicine, University of Cologne, Germany; German Research Center for Neurodegenerative Diseases (DZNE), Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, University of Cologne, Germany; German Research Center for Neurodegenerative Diseases (DZNE), Germany.
| |
Collapse
|
34
|
Choi Y, Ha S, Lee YS, Kim YK, Lee DS, Kim DJ. Development of tau PET Imaging Ligands and their Utility in Preclinical and Clinical Studies. Nucl Med Mol Imaging 2017; 52:24-30. [PMID: 29391909 DOI: 10.1007/s13139-017-0484-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/10/2017] [Accepted: 05/22/2017] [Indexed: 12/16/2022] Open
Abstract
The pathological features of Alzheimer's disease are senile plaques which are aggregates of β-amyloid peptides and neurofibrillary tangles in the brain. Neurofibrillary tangles are aggregates of hyperphosphorylated tau proteins, and these induce various other neurodegenerative diseases, such as progressive supranuclear palsy, corticobasal degeneration, frontotemporal lobar degeneration, frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17), and chronic traumatic encephalopathy. In the case of Alzheimer's disease, the measurement of neurofibrillary tangles associated with cognitive decline is suitable for differential diagnosis, disease progression assessment, and to monitor the effects of therapeutic treatment. This review discusses considerations for the development of tau ligands for imaging and summarizes the results of the first-in-human and preclinical studies of the tau tracers that have been developed thus far. The development of tau ligands for imaging studies will be helpful for differential diagnosis and for the development of therapeutic treatments for tauopathies including Alzheimer's disease.
Collapse
Affiliation(s)
- Yoori Choi
- 1Department of Nuclear Medicine, College of Medicine, Seoul National University, 110-744, 28 Yongon-Dong, Jongno-Gu, Seoul, South Korea.,2Department of Nuclear Medicine, Seoul National University Hospital, 28 Yongon-Dong, Jongno-Gu, Seoul, 110-744 South Korea
| | - Seunggyun Ha
- 1Department of Nuclear Medicine, College of Medicine, Seoul National University, 110-744, 28 Yongon-Dong, Jongno-Gu, Seoul, South Korea.,3Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, 03080, 103 Daehak-ro, Jongno-gu, Seoul, South Korea
| | - Yun-Sang Lee
- 1Department of Nuclear Medicine, College of Medicine, Seoul National University, 110-744, 28 Yongon-Dong, Jongno-Gu, Seoul, South Korea.,3Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, 03080, 103 Daehak-ro, Jongno-gu, Seoul, South Korea
| | - Yun Kyung Kim
- 4Institute of Brain Science, Korean Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 136-791 South Korea
| | - Dong Soo Lee
- 1Department of Nuclear Medicine, College of Medicine, Seoul National University, 110-744, 28 Yongon-Dong, Jongno-Gu, Seoul, South Korea.,2Department of Nuclear Medicine, Seoul National University Hospital, 28 Yongon-Dong, Jongno-Gu, Seoul, 110-744 South Korea.,3Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, 03080, 103 Daehak-ro, Jongno-gu, Seoul, South Korea
| | - Dong Jin Kim
- 4Institute of Brain Science, Korean Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 136-791 South Korea
| |
Collapse
|
35
|
Saint-Aubert L, Lemoine L, Chiotis K, Leuzy A, Rodriguez-Vieitez E, Nordberg A. Tau PET imaging: present and future directions. Mol Neurodegener 2017; 12:19. [PMID: 28219440 PMCID: PMC5319037 DOI: 10.1186/s13024-017-0162-3] [Citation(s) in RCA: 210] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 02/15/2017] [Indexed: 12/15/2022] Open
Abstract
Abnormal aggregation of tau in the brain is a major contributing factor in various neurodegenerative diseases. The role of tau phosphorylation in the pathophysiology of tauopathies remains unclear. Consequently, it is important to be able to accurately and specifically target tau deposits in vivo in the brains of patients. The advances of molecular imaging in the recent years have now led to the recent development of promising tau-specific tracers for positron emission tomography (PET), such as THK5317, THK5351, AV-1451, and PBB3. These tracers are now available for clinical assessment in patients with various tauopathies, including Alzheimer's disease, as well as in healthy subjects. Exploring the patterns of tau deposition in vivo for different pathologies will allow discrimination between neurodegenerative diseases, including different tauopathies, and monitoring of disease progression. The variety and complexity of the different types of tau deposits in the different diseases, however, has resulted in quite a challenge for the development of tau PET tracers. Extensive work remains in order to fully characterize the binding properties of the tau PET tracers, and to assess their usefulness as an early biomarker of the underlying pathology. In this review, we summarize recent findings on the most promising tau PET tracers to date, discuss what has been learnt from these findings, and offer some suggestions for the next steps that need to be achieved in a near future.
Collapse
Affiliation(s)
- Laure Saint-Aubert
- Department NVS, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Novum 5th floor, 141 57, Huddinge, Sweden
| | - Laetitia Lemoine
- Department NVS, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Novum 5th floor, 141 57, Huddinge, Sweden
| | - Konstantinos Chiotis
- Department NVS, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Novum 5th floor, 141 57, Huddinge, Sweden
| | - Antoine Leuzy
- Department NVS, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Novum 5th floor, 141 57, Huddinge, Sweden
| | - Elena Rodriguez-Vieitez
- Department NVS, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Novum 5th floor, 141 57, Huddinge, Sweden
| | - Agneta Nordberg
- Department NVS, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Novum 5th floor, 141 57, Huddinge, Sweden. .,Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|