1
|
Kong Y, Maschio CA, Shi X, Xie F, Zuo C, Konietzko U, Shi K, Rominger A, Xiao J, Huang Q, Nitsch RM, Guan Y, Ni R. Relationship Between Reactive Astrocytes, by [ 18F]SMBT-1 Imaging, with Amyloid-Beta, Tau, Glucose Metabolism, and TSPO in Mouse Models of Alzheimer's Disease. Mol Neurobiol 2024; 61:8387-8401. [PMID: 38502413 DOI: 10.1007/s12035-024-04106-7] [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: 09/25/2023] [Accepted: 03/06/2024] [Indexed: 03/21/2024]
Abstract
Reactive astrocytes play an important role in the development of Alzheimer's disease (AD). Here, we aimed to investigate the temporospatial relationships among monoamine oxidase-B, tau and amyloid-β (Aβ), translocator protein, and glucose metabolism by using multitracer imaging in AD transgenic mouse models. Positron emission tomography (PET) imaging with [18F]SMBT-1 (monoamine oxidase-B), [18F]florbetapir (Aβ), [18F]PM-PBB3 (tau), [18F]fluorodeoxyglucose (FDG), and [18F]DPA-714 (translocator protein) was carried out in 5- and 10-month-old APP/PS1, 11-month-old 3×Tg mice, and aged-matched wild-type mice. The brain regional referenced standard uptake value (SUVR) was computed with the cerebellum as the reference region. Immunofluorescence staining was performed on mouse brain tissue slices. [18F]SMBT-1 and [18F]florbetapir SUVRs were greater in the cortex and hippocampus of 10-month-old APP/PS1 mice than in those of 5-month-old APP/PS1 mice and wild-type mice. No significant difference in the regional [18F]FDG or [18F]DPA-714 SUVRs was observed in the brains of 5- or 10-month-old APP/PS1 mice or wild-type mice. No significant difference in the SUVRs of any tracer was observed between 11-month-old 3×Tg mice and age-matched wild-type mice. A positive correlation between the SUVRs of [18F]florbetapir and [18F]DPA-714 in the cortex and hippocampus was observed among the transgenic mice. Immunostaining validated the distribution of MAO-B and limited Aβ and tau pathology in 11-month-old 3×Tg mice; and Aβ deposits in brain tissue from 10-month-old APP/PS1 mice. In summary, these findings provide in vivo evidence that an increase in astrocyte [18F]SMBT-1 accompanies Aβ accumulation in APP/PS1 models of AD amyloidosis.
Collapse
Affiliation(s)
- Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Cinzia A Maschio
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Zurich Neuroscience Zentrum (ZNZ), Zurich, Switzerland
| | - Xuefeng Shi
- Qinghai Provincial People's Hospital, Xining, China
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuantao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Uwe Konietzko
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland
| | - Jianfei Xiao
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qi Huang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Roger M Nitsch
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China.
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.
- Zurich Neuroscience Zentrum (ZNZ), Zurich, Switzerland.
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland.
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
2
|
Sison SA, Paclibar CG, Liang C, Mukherjee J. Radioiodinated Tau Imaging Agent III Molecular Modeling, Synthesis, and Evaluation of a New Tau Imaging Agent, [ 125I]ISAS in Post-Mortem Human Alzheimer's Disease Brain. Molecules 2024; 29:3308. [PMID: 39064887 PMCID: PMC11279437 DOI: 10.3390/molecules29143308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Using a molecular modeling approach for Tau-binding sites, we modified our previously reported imaging agent, [125I]INFT, for the potential improvement of binding properties to Tau in an Alzheimer's disease (AD) brain. Two new derivatives, namely [125I]ISAS and [125I]NIPZ, were designed, where binding energies at site 1 of Tau were -7.4 and -6.0 kcal/mole, respectively, compared to [125I]INFT (-7.6 kcal/mole). The radiosynthesis of [125I]ISAS and [125I]NIPZ was carried out by using iodine-125 and purified chromatographically to achieve >90% purity. In vitro binding affinities (IC50) for Tau were as follows: INFT = 7.3 × 10-8 M; ISAS = 4.7 × 10-8 M; NIPZ > 10-6 M. The binding of [125I]ISAS to gray matter (GM) correlated with the presence of Tau in the AD brain, confirmed by anti-Tau immunohistochemistry. [125I]NIPZ did not bind to Tau, with similar levels of binding observed in GM and white matter (WM). Four radiotracers were compared and the rank order of binding to Tau was found to be [125I]IPPI > [125I]INFT > [125I]ISAS >>> [125I]NIPZ with GM/WM ratios of [125I]IPPI = 7.74 > [125I]INFT = 4.86 > [125I]ISAS = 3.62 >> [125I]NIPZ = 1.24. The predictive value of Chimera-AutoDock for structurally related compounds binding to the Tau binding sites (measured as binding energy) was good. A binding energy of less than -7 kcal/mole is necessary and less than -8 kcal/mole will be more suitable for developing imaging agents.
Collapse
Affiliation(s)
| | | | | | - Jogeshwar Mukherjee
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA; (S.A.S.); (C.G.P.); (C.L.)
| |
Collapse
|
3
|
Espargaró A, Álvarez-Berbel I, Busquets MA, Sabate R. In Vivo Assays for Amyloid-Related Diseases. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2024; 17:433-458. [PMID: 38598824 DOI: 10.1146/annurev-anchem-061622-023326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Amyloid-related diseases, such as Alzheimer's and Parkinson's disease, are devastating conditions caused by the accumulation of abnormal protein aggregates known as amyloid fibrils. While assays involving animal models are essential for understanding the pathogenesis and developing therapies, a wide array of standard analytical techniques exists to enhance our understanding of these disorders. These techniques provide valuable information on the formation and propagation of amyloid fibrils, as well as the pharmacokinetics and pharmacodynamics of candidate drugs. Despite ethical concerns surrounding animal use, animal models remain vital tools in the search for treatments. Regardless of the specific animal model chosen, the analytical methods used are usually standardized. Therefore, the main objective of this review is to categorize and outline the primary analytical methods used in in vivo assays for amyloid-related diseases, highlighting their critical role in furthering our understanding of these disorders and developing effective therapies.
Collapse
Affiliation(s)
- Alba Espargaró
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
- 2Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Irene Álvarez-Berbel
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
| | - Maria Antònia Busquets
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
- 2Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Raimon Sabate
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
- 2Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| |
Collapse
|
4
|
Fernandez ME, Martinez-Romero J, Aon MA, Bernier M, Price NL, de Cabo R. How is Big Data reshaping preclinical aging research? Lab Anim (NY) 2023; 52:289-314. [PMID: 38017182 DOI: 10.1038/s41684-023-01286-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 10/10/2023] [Indexed: 11/30/2023]
Abstract
The exponential scientific and technological progress during the past 30 years has favored the comprehensive characterization of aging processes with their multivariate nature, leading to the advent of Big Data in preclinical aging research. Spanning from molecular omics to organism-level deep phenotyping, Big Data demands large computational resources for storage and analysis, as well as new analytical tools and conceptual frameworks to gain novel insights leading to discovery. Systems biology has emerged as a paradigm that utilizes Big Data to gain insightful information enabling a better understanding of living organisms, visualized as multilayered networks of interacting molecules, cells, tissues and organs at different spatiotemporal scales. In this framework, where aging, health and disease represent emergent states from an evolving dynamic complex system, context given by, for example, strain, sex and feeding times, becomes paramount for defining the biological trajectory of an organism. Using bioinformatics and artificial intelligence, the systems biology approach is leading to remarkable advances in our understanding of the underlying mechanism of aging biology and assisting in creative experimental study designs in animal models. Future in-depth knowledge acquisition will depend on the ability to fully integrate information from different spatiotemporal scales in organisms, which will probably require the adoption of theories and methods from the field of complex systems. Here we review state-of-the-art approaches in preclinical research, with a focus on rodent models, that are leading to conceptual and/or technical advances in leveraging Big Data to understand basic aging biology and its full translational potential.
Collapse
Affiliation(s)
- Maria Emilia Fernandez
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Jorge Martinez-Romero
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Miguel A Aon
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Michel Bernier
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Nathan L Price
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Rafael de Cabo
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
5
|
Limpengco RR, Liang C, Sandhu YK, Mukherjee J. [ 125I]INFT: Synthesis and Evaluation of a New Imaging Agent for Tau Protein in Post-Mortem Human Alzheimer's Disease Brain. Molecules 2023; 28:5769. [PMID: 37570739 PMCID: PMC10421386 DOI: 10.3390/molecules28155769] [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: 06/03/2023] [Revised: 07/24/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Aggregation of Tau protein into paired helical filaments causing neurofibrillary tangles (NFT) is a neuropathological feature in Alzheimer's disease (AD). This study aimed to develop and evaluate the effectiveness of a novel radioiodinated tracer, 4-[125I]iodo-3-(1H-pyrrolo[2,3-c]pyridine-1-yl)pyridine ([125I]INFT), for binding to Tau protein in postmortem human AD brain. Radiosynthesis of [125I]INFT was carried out using electrophilic destannylation by iodine-125 and purified chromatographically. Computational modeling of INFT binding on Tau fibril was compared with IPPI. In vitro, autoradiography studies were conducted with [125I]INFT for Tau in AD and cognitively normal (CN) brains. [125I]INFT was produced in >95% purity. Molecular modeling of INFT revealed comparable binding energies to IPPI at site-1 of the Tau fibril with an affinity of IC50 = 7.3 × 10-8 M. Binding of [125I]INFT correlated with the presence of Tau in the AD brain, confirmed by anti-Tau immunohistochemistry. The ratio of average grey matter (GM) [125I]INFT in AD versus CN was found to be 5.9, and AD GM/white matter (WM) = 2.5. Specifically bound [125I]INFT to Tau in AD brains was displaced by IPPI (>90%). Monoamine oxidase inhibitor deprenyl had no effect and clorgyline had little effect on [125I]INFT binding. [125I]INFT is a less lipophilic imaging agent for Tau in AD.
Collapse
Affiliation(s)
- Roz R Limpengco
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA
| | - Christopher Liang
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA
| | - Yasmin K Sandhu
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA
| | - Jogeshwar Mukherjee
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA
| |
Collapse
|
6
|
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
|
7
|
Lai C, Chen Z, Ding Y, Chen Q, Su S, Liu H, Ni R, Tang Z. Rapamycin Attenuated Zinc-Induced Tau Phosphorylation and Oxidative Stress in Rats: Involvement of Dual mTOR/p70S6K and Nrf2/HO-1 Pathways. Front Immunol 2022; 13:782434. [PMID: 35197970 PMCID: PMC8858937 DOI: 10.3389/fimmu.2022.782434] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 01/13/2022] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease is pathologically characterized by abnormal accumulation of amyloid-beta plaques, neurofibrillary tangles, oxidative stress, neuroinflammation, and neurodegeneration. Metal dysregulation, including excessive zinc released by presynaptic neurons, plays an important role in tau pathology and oxidase activation. The activities of mammalian target of rapamycin (mTOR)/ribosomal S6 protein kinase (p70S6K) are elevated in the brains of patients with Alzheimer's disease. Zinc induces tau hyperphosphorylation via mTOR/P70S6K activation in vitro. However, the involvement of the mTOR/P70S6K pathway in zinc-induced oxidative stress, tau degeneration, and synaptic and cognitive impairment has not been fully elucidated in vivo. Here, we assessed the effect of pathological zinc concentrations in SH-SY5Y cells by using biochemical assays and immunofluorescence staining. Rats (n = 18, male) were laterally ventricularly injected with zinc, treated with rapamycin (intraperitoneal injection) for 1 week, and assessed using the Morris water maze. Evaluation of oxidative stress, tau phosphorylation, and synaptic impairment was performed using the hippocampal tissue of the rats by biochemical assays and immunofluorescence staining. The results from the Morris water maze showed that the capacity of spatial memory was impaired in zinc-treated rats. Zinc sulfate significantly increased the levels of P-mTOR Ser2448, P-p70S6K Thr389, and P-tau Ser356 and decreased the levels of nuclear factor erythroid 2-related factor-2 (Nrf2) and heme oxygenase-1 (HO-1) in SH-SY5Y cells and in zinc-treated rats compared with the control groups. Increased expression of reactive oxygen species was observed in zinc sulfate-induced SH-SY5Y cells and in the hippocampus of zinc-injected rats. Rapamycin, an inhibitor of mTOR, rescued zinc-induced increases in mTOR/p70S6K activation, tau phosphorylation, and oxidative stress, and Nrf2/HO-1 inactivation, cognitive impairment, and synaptic impairment reduced the expression of synapse-related proteins in zinc-injected rats. In conclusion, our findings imply that rapamycin prevents zinc-induced cognitive impairment and protects neurons from tau pathology, oxidative stress, and synaptic impairment by decreasing mTOR/p70S6K hyperactivity and increasing Nrf2/HO-1 activity.
Collapse
Affiliation(s)
- Chencen Lai
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, China
- Department of Nosocomial Infection, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Zhuyi Chen
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, China
| | - Yuanting Ding
- Department of Nosocomial Infection, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Qian Chen
- Department of Nosocomial Infection, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Songbai Su
- Department of Nosocomial Infection, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Heng Liu
- Department of Anesthesiology, Tongren Municipal People’s Hospital, Tongren, China
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, Eidgenössische Technische Hochschule Zürich (ETH) and University of Zurich, Zurich, Switzerland
| | - Zhi Tang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, China
| |
Collapse
|
8
|
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
|