1
|
Chauveau F, Winkeler A, Chalon S, Boutin H, Becker G. PET imaging of neuroinflammation: any credible alternatives to TSPO yet? Mol Psychiatry 2025; 30:213-228. [PMID: 38997465 DOI: 10.1038/s41380-024-02656-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024]
Abstract
Over the last decades, the role of neuroinflammation in neuropsychiatric conditions has attracted an exponentially growing interest. A key driver for this trend was the ability to image brain inflammation in vivo using PET radioligands targeting the Translocator Protein 18 kDa (TSPO), which is known to be expressed in activated microglia and astrocytes upon inflammatory events as well as constitutively in endothelial cells. TSPO is a mitochondrial protein that is expressed mostly by microglial cells upon activation but is also expressed by astrocytes in some conditions and constitutively by endothelial cells. Therefore, our current understanding of neuroinflammation dynamics is hampered by the lack of alternative targets available for PET imaging. We performed a systematic search and review on radiotracers developed for neuroinflammation PET imaging apart from TSPO. The following targets of interest were identified through literature screening (including previous narrative reviews): P2Y12R, P2X7R, CSF1R, COX (microglial targets), MAO-B, I2BS (astrocytic targets), CB2R & S1PRs (not specific of a single cell type). We determined the level of development and provided a scoping review for each target. Strikingly, astrocytic biomarker MAO-B has progressed in clinical investigations the furthest, while few radiotracers (notably targeting S1P1Rs, CSF1R) are being implemented in clinical investigations. Other targets such as CB2R and P2X7R have proven disappointing in clinical studies (e.g. poor signal, lack of changes in disease conditions, etc.). While astrocytic targets are promising, development of new biomarkers and tracers specific for microglial activation has proven challenging.
Collapse
Affiliation(s)
- Fabien Chauveau
- Université Claude Bernard Lyon 1, Centre de Recherche en Neurosciences de Lyon, Inserm U1028, CNRS UMR5292, BIORAN, Groupement Hospitalier Est - CERMEP, 59 boulevard Pinel, 69677, Bron, Cedex, France
| | - Alexandra Winkeler
- Université Paris-Saclay, Inserm, CNRS, CEA, BioMaps, Service Hospitalier Frédéric Joliot, 4 place du général Leclerc, 91401, Orsay, France
| | - Sylvie Chalon
- UMR 1253 iBrain, Université de Tours - INSERM, Bâtiment Planiol, UFR de Médecine, 10 Boulevard Tonnellé, 37032, Tours, Cedex 01, France
| | - Hervé Boutin
- UMR 1253 iBrain, Université de Tours - INSERM, Bâtiment Planiol, UFR de Médecine, 10 Boulevard Tonnellé, 37032, Tours, Cedex 01, France.
| | - Guillaume Becker
- Université Claude Bernard Lyon 1, Centre de Recherche en Neurosciences de Lyon, Inserm U1028, CNRS UMR5292, BIORAN, Groupement Hospitalier Est - CERMEP, 59 boulevard Pinel, 69677, Bron, Cedex, France
- Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail, 14 rue Pierre et Marie Curie, 94701, Maisons-Alfort, Cedex, France
| |
Collapse
|
2
|
Chongtham A, Sharma A, Nath B, Murtha K, Gorbachev K, Ramakrishnan A, Schmidt EF, Shen L, Pereira AC. Common and divergent pathways in early stages of glutamate and tau-mediated toxicities in neurodegeneration. Exp Neurol 2024; 382:114967. [PMID: 39326823 DOI: 10.1016/j.expneurol.2024.114967] [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: 05/02/2024] [Revised: 09/17/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024]
Abstract
It has been shown that excitotoxicity and tau-mediated toxicities are major contributing factors to neuronal death in Alzheimer's disease (AD). The excitatory amino acid transporter 2 (EAAT2 or GLT-1), the major glutamate transporter in the brain that regulates glutamate levels synaptically and extrasynaptically, has been shown to be deficient in AD brains, leading to excitotoxicity and subsequent cell death. Similarly, buildup of neurofibrillary tangles, which consist of hyperphosphorylated tau protein, correlates with cognitive decline and neuronal atrophy in AD. However, common genes and pathways that are critical in the aforementioned toxicities have not been well elucidated. To investigate the impact of glutamate dyshomeostasis and tau accumulation on translational profiles of affected hippocampal neurons, we used mouse models of excitotoxicity and tau-mediated toxicities (GLT-1-/- and P301S, respectively) in conjunction with BAC-TRAP technology. Our data show that GLT-1 deficiency in CA3 pyramidal neurons leads to translational signatures characterized by dysregulation of pathways associated with synaptic plasticity and neuronal survival, while the P301S mutation induces changes in endocytic pathways and mitochondrial dysfunction. Finally, the commonly dysregulated pathways include impaired ion homeostasis and metabolic pathways. These common pathways may shed light on potential therapeutic targets for ameliorating glutamate and tau-mediated toxicities in AD.
Collapse
Affiliation(s)
- Anjalika Chongtham
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Abhijeet Sharma
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Banshi Nath
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Kaitlin Murtha
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Kirill Gorbachev
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Aarthi Ramakrishnan
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Eric F Schmidt
- The Rockefeller University, Laboratory of Molecular Biology, New York, NY 10065, United States of America
| | - Li Shen
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Ana C Pereira
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America.
| |
Collapse
|
3
|
Lopresti BJ, Stehouwer J, Reese AC, Mason NS, Royse SK, Narendran R, Laymon CM, Lopez OL, Cohen AD, Mathis CA, Villemagne VL. Kinetic modeling of the monoamine oxidase-B radioligand [ 18F]SMBT-1 in human brain with positron emission tomography. J Cereb Blood Flow Metab 2024; 44:1262-1276. [PMID: 38735059 PMCID: PMC11542143 DOI: 10.1177/0271678x241254679] [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: 10/19/2023] [Revised: 02/29/2024] [Accepted: 04/07/2024] [Indexed: 05/14/2024]
Abstract
This paper describes pharmacokinetic analyses of the monoamine-oxidase-B (MAO-B) radiotracer [18F](S)-(2-methylpyrid-5-yl)-6-[(3-fluoro-2-hydroxy)propoxy]quinoline ([18F]SMBT-1) for positron emission tomography (PET) brain imaging. Brain MAO-B expression is widespread, predominantly within astrocytes. Reactive astrogliosis in response to neurodegenerative disease pathology is associated with MAO-B overexpression. Fourteen elderly subjects (8 control, 5 mild cognitive impairment, 1 Alzheimer's disease) with amyloid ([11C]PiB) and tau ([18F]flortaucipir) imaging assessments underwent dynamic [18F]SMBT-1 PET imaging with arterial input function determination. [18F]SMBT-1 showed high brain uptake and a retention pattern consistent with the known MAO-B distribution. A two-tissue compartment (2TC) model where the K1/k2 ratio was fixed to a whole brain value best described [18F]SMBT-1 kinetics. The 2TC total volume of distribution (VT) was well identified and highly correlated (r2∼0.8) with post-mortem MAO-B indices. Cerebellar grey matter (CGM) showed the lowest mean VT of any region and is considered the optimal pseudo-reference region. Simplified analysis methods including reference tissue models, non-compartmental models, and standard uptake value ratios (SUVR) agreed with 2TC outcomes (r2 > 0.9) but with varying bias. We found the CGM-normalized 70-90 min SUVR to be highly correlated (r2 = 0.93) with the 2TC distribution volume ratio (DVR) with acceptable bias (∼10%), representing a practical alternative for [18F]SMBT-1 analyses.
Collapse
Affiliation(s)
- Brian J Lopresti
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jeffrey Stehouwer
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alexandria C Reese
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Neale S Mason
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sarah K Royse
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rajesh Narendran
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Charles M Laymon
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Dept. of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Oscar L Lopez
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Clinical and Translational Sciences Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ann D Cohen
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Victor L Villemagne
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Corriveau-Lecavalier N, Adams JN, Fischer L, Molloy EN, Maass A. Cerebral hyperactivation across the Alzheimer's disease pathological cascade. Brain Commun 2024; 6:fcae376. [PMID: 39513091 PMCID: PMC11542485 DOI: 10.1093/braincomms/fcae376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/18/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024] Open
Abstract
Neuronal dysfunction in specific brain regions or across distributed brain networks is a known feature of Alzheimer's disease. An often reported finding in the early stage of the disease is the presence of increased functional MRI (fMRI) blood oxygenation level-dependent signal under task conditions relative to cognitively normal controls, a phenomenon known as 'hyperactivation'. However, research in the past decades yielded complex, sometimes conflicting results. The magnitude and topology of fMRI hyperactivation patterns have been found to vary across the preclinical and clinical spectrum of Alzheimer's disease, including concomitant 'hypoactivation' in some cases. These incongruences are likely due to a range of factors, including the disease stage at which the cohort is examined, the brain areas or networks studied and the fMRI paradigm utilized to evoke these functional abnormalities. Additionally, a perennial question pertains to the nature of hyperactivation in the context of Alzheimer's disease. Some propose it reflects compensatory mechanisms to sustain cognitive performance, while others suggest it is linked to the pathological disruption of a highly regulated homeostatic cycle that contributes to, or even drives, disease progression. Providing a coherent narrative for these empirical and conceptual discrepancies is paramount to develop disease models, understand the synergy between hyperactivation and the Alzheimer's disease pathological cascade and tailor effective interventions. We first provide a comprehensive overview of functional brain changes spanning the course from normal ageing to the clinical spectrum of Alzheimer's disease. We then highlight evidence supporting a close relationship between fMRI hyperactivation and in vivo markers of Alzheimer's pathology. We primarily focus on task-based fMRI studies in humans, but also consider studies using different functional imaging techniques and animal models. We then discuss the potential mechanisms underlying hyperactivation in the context of Alzheimer's disease and provide a testable framework bridging hyperactivation, ageing, cognition and the Alzheimer's disease pathological cascade. We conclude with a discussion of future challenges and opportunities to advance our understanding of the fundamental disease mechanisms of Alzheimer's disease, and the promising development of therapeutic interventions incorporating or aimed at hyperactivation and large-scale functional systems.
Collapse
Affiliation(s)
- Nick Corriveau-Lecavalier
- Department of Neurology, Mayo Clinic, Rochester, Minnesota 55902, USA
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota 55902 USA
| | - Jenna N Adams
- Department of Neurobiology and Behavior, University of California, Irvine 92697, CA, USA
| | - Larissa Fischer
- German Center for Neurodegenerative Diseases, Magdeburg 39120, Germany
| | - Eóin N Molloy
- German Center for Neurodegenerative Diseases, Magdeburg 39120, Germany
- Division of Nuclear Medicine, Department of Radiology & Nuclear Medicine, Faculty of Medicine, Otto von Guericke University Magdeburg, Magdeburg 39120, Germany
| | - Anne Maass
- German Center for Neurodegenerative Diseases, Magdeburg 39120, Germany
- Institute for Biology, Otto-von-Guericke University Magdeburg, Magdeburg 39120, Germany
| |
Collapse
|
5
|
Nam MH, Na H, Justin Lee C, Yun M. A Key Mediator and Imaging Target in Alzheimer's Disease: Unlocking the Role of Reactive Astrogliosis Through MAOB. Nucl Med Mol Imaging 2024; 58:177-184. [PMID: 38932762 PMCID: PMC11196512 DOI: 10.1007/s13139-023-00837-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/18/2023] [Accepted: 12/26/2023] [Indexed: 06/28/2024] Open
Abstract
Astrocytes primarily maintain physiological brain homeostasis. However, under various pathological conditions, they can undergo morphological, transcriptomic, and functional transformations, collectively referred to as reactive astrogliosis. Recent studies have accumulated lines of evidence that reactive astrogliosis plays a crucial role in the pathology of Alzheimer's disease (AD). In particular, monoamine oxidase B, a mitochondrial enzyme mainly expressed in astrocytes, significantly contributes to neuronal dysfunction and neurodegeneration in AD brains. Moreover, it has been reported that reactive astrogliosis precedes other pathological hallmarks such as amyloid-beta plaque deposition and tau tangle formation in AD. Due to the early onset and profound impact of reactive astrocytes on pathology, there have been extensive efforts in the past decade to visualize these cells in the brains of AD patients using positron emission tomography (PET) imaging. In this review, we summarize the recent studies regarding the essential pathological importance of reactive astrocytes in AD and their application as a target for PET imaging.
Collapse
Affiliation(s)
- Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Heesu Na
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - C. Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Mijin Yun
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
6
|
Edison P. Astroglial activation: Current concepts and future directions. Alzheimers Dement 2024; 20:3034-3053. [PMID: 38305570 PMCID: PMC11032537 DOI: 10.1002/alz.13678] [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: 08/24/2023] [Revised: 11/27/2023] [Accepted: 12/11/2023] [Indexed: 02/03/2024]
Abstract
Astrocytes are abundantly and ubiquitously expressed cell types with diverse functions throughout the central nervous system. Astrocytes show remarkable plasticity and exhibit morphological, molecular, and functional remodeling in response to injury, disease, or infection of the central nervous system, as evident in neurodegenerative diseases. Astroglial mediated inflammation plays a prominent role in the pathogenesis of neurodegenerative diseases. This review focus on the role of astrocytes as essential players in neuroinflammation and discuss their morphological and functional heterogeneity in the normal central nervous system and explore the spatial and temporal variations in astroglial phenotypes observed under different disease conditions. This review discusses the intimate relationship of astrocytes to pathological hallmarks of neurodegenerative diseases. Finally, this review considers the putative therapeutic strategies that can be deployed to modulate the astroglial functions in neurodegenerative diseases. HIGHLIGHTS: Astroglia mediated neuroinflammation plays a key role in the pathogenesis of neurodegenerative diseases. Activated astrocytes exhibit diverse phenotypes in a region-specific manner in brain and interact with β-amyloid, tau, and α-synuclein species as well as with microglia and neuronal circuits. Activated astrocytes are likely to influence the trajectory of disease progression of neurodegenerative diseases, as determined by the stage of disease, individual susceptibility, and state of astroglial priming. Modulation of astroglial activation may be a therapeutic strategy at various stages in the trajectory of neurodegenerative diseases to modify the disease course.
Collapse
Affiliation(s)
- Paul Edison
- Division of NeurologyDepartment of Brain SciencesFaculty of Medicine, Imperial College LondonLondonUK
- Division of Psychological medicine and clinical neurosciencesSchool of Medicine, Cardiff UniversityWalesUK
| |
Collapse
|
7
|
Rodriguez-Vieitez E, Kumar A, Malarte ML, Ioannou K, Rocha FM, Chiotis K. Imaging Neuroinflammation: Quantification of Astrocytosis in a Multitracer PET Approach. Methods Mol Biol 2024; 2785:195-218. [PMID: 38427196 DOI: 10.1007/978-1-0716-3774-6_13] [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] [Indexed: 03/02/2024]
Abstract
The recent progress in the development of in vivo biomarkers is rapidly changing how neurodegenerative diseases are conceptualized and diagnosed and how clinical trials are designed today. Alzheimer's disease (AD) - the most common neurodegenerative disorder - is characterized by a complex neuropathology involving the deposition of extracellular amyloid-β (Aβ) plaques and intracellular neurofibrillary tangles (NFTs) of hyperphosphorylated tau proteins, accompanied by the activation of glial cells, i.e., astrocytes and microglia, and neuroinflammatory response, leading to neurodegeneration and cognitive dysfunction. An increasing diversity of positron emission tomography (PET) imaging radiotracers is available to selectively target the different pathophysiological processes of AD. Along with the success of Aβ PET and the more recent tau PET imaging, there is a great interest to develop PET tracers to image glial reactivity and neuroinflammation. While most research to date has focused on imaging microgliosis, there is an upsurge of interest in imaging reactive astrocytes in the AD continuum. There is increasing evidence that reactive astrocytes are morphologically and functionally heterogeneous, with different subtypes that express different markers and display various homeostatic or detrimental roles across disease stages. Therefore, multiple biomarkers are desirable to unravel the complex phenomenon of reactive astrocytosis. In the field of in vivo PET imaging in AD, the research concerning reactive astrocytes has predominantly focused on targeting monoamine oxidase B (MAO-B), most often using either 11C-deuterium-L-deprenyl (11C-DED) or 18F-SMBT-1 PET tracers. Additionally, imidazoline2 binding (I2BS) sites have been imaged using 11C-BU99008 PET. Recent studies in our group using 11C-DED PET imaging suggest that astrocytosis may be present from the early stages of disease development in AD. This chapter provides a detailed description of the practical approach used for the analysis of 11C-DED PET imaging data in a multitracer PET paradigm including 11C-Pittsburgh compound B (11C-PiB) and 18F-fluorodeoxyglucose (18F-FDG). The multitracer PET approach allows investigating the comparative regional and temporal patterns of in vivo brain astrocytosis, fibrillar Aβ deposition, glucose metabolism, and brain structural changes. It may also contribute to understanding the potential role of novel plasma biomarkers of reactive astrocytes, in particular the glial fibrillary acidic protein (GFAP), at different stages of disease progression. This chapter attempts to stimulate further research in the field, including the development of novel PET tracers that may allow visualizing different aspects of the complex astrocytic and microglial response in neurodegenerative diseases. Progress in the field will contribute to the incorporation of PET imaging of glial reactivity and neuroinflammation as biomarkers with clinical application and motivate further investigation on glial cells as therapeutic targets in AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Elena Rodriguez-Vieitez
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| | - Amit Kumar
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Mona-Lisa Malarte
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Konstantinos Ioannou
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Filipa M Rocha
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Konstantinos Chiotis
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
8
|
Wang YTT, Rosa-Neto P, Gauthier S. Advanced brain imaging for the diagnosis of Alzheimer disease. Curr Opin Neurol 2023; 36:481-490. [PMID: 37639461 DOI: 10.1097/wco.0000000000001198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
PURPOSE OF REVIEW The purpose is to review the latest advances of brain imaging for the diagnosis of Alzheimer disease (AD). RECENT FINDINGS Brain imaging techniques provide valuable and complementary information to support the diagnosis of Alzheimer disease in clinical and research settings. The recent FDA accelerated approvals of aducanumab, lecanemab and donanemab made amyloid-PET critical in helping determine the optimal window for anti-amyloid therapeutic interventions. Tau-PET, on the other hand, is considered of key importance for the tracking of disease progression and for monitoring therapeutic interventions in clinical trials. PET imaging for microglial activation, astrocyte reactivity and synaptic degeneration are still new techniques only used in the research field, and more studies are needed to validate their use in the clinical diagnosis of AD. Finally, artificial intelligence has opened new prospective in the early detection of AD using MRI modalities. SUMMARY Brain imaging techniques using PET improve our understanding of the different AD-related pathologies and their relationship with each other along the course of disease. With more robust validation, machine learning and deep learning algorithms could be integrated with neuroimaging modalities to serve as valuable tools for clinicians to make early diagnosis and prognosis of AD.
Collapse
|
9
|
Chiotis K, Johansson C, Rodriguez-Vieitez E, Ashton NJ, Blennow K, Zetterberg H, Graff C, Nordberg A. Tracking reactive astrogliosis in autosomal dominant and sporadic Alzheimer's disease with multi-modal PET and plasma GFAP. Mol Neurodegener 2023; 18:60. [PMID: 37697307 PMCID: PMC10496408 DOI: 10.1186/s13024-023-00647-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 08/07/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND Plasma assays for the detection of Alzheimer's disease neuropathological changes are receiving ever increasing interest. The concentration of plasma glial fibrillary acidic protein (GFAP) has been suggested as a potential marker of astrocytes or recently, amyloid-β burden, although this hypothesis remains unproven. We compared plasma GFAP levels with the astrocyte tracer 11C-Deuterium-L-Deprenyl (11C-DED) in a multi-modal PET design in participants with sporadic and Autosomal Dominant Alzheimer's disease. METHODS Twenty-four individuals from families with known Autosomal Dominant Alzheimer's Disease mutations (mutation carriers = 10; non-carriers = 14) and fifteen patients with sporadic Alzheimer's disease were included. The individuals underwent PET imaging with 11C-DED, 11C-PIB and 18F-FDG, as markers of reactive astrogliosis, amyloid-β deposition, and glucose metabolism, respectively, and plasma sampling for measuring GFAP concentrations. Twenty-one participants from the Autosomal Dominant Alzheimer's Disease group underwent follow-up plasma sampling and ten of these participants underwent follow-up PET imaging. RESULTS In mutation carriers, plasma GFAP levels and 11C-PIB binding increased, while 11C-DED binding and 18F-FDG uptake significantly decreased across the estimated years to symptom onset. Cross-sectionally, plasma GFAP demonstrated a negative correlation with 11C-DED binding in both mutation carriers and patients with sporadic disease. Plasma GFAP indicated cross-sectionally a significant positive correlation with 11C-PIB binding and a significant negative correlation with 18F-FDG in the whole sample. The longitudinal levels of 11C-DED binding showed a significant negative correlation with longitudinal plasma GFAP concentrations over the follow-up interval. CONCLUSIONS Plasma GFAP concentration and astrocyte 11C-DED brain binding levels followed divergent trajectories and may reflect different underlying processes. The strong negative association between plasma GFAP and 11C-DED binding in Autosomal Dominant and sporadic Alzheimer's disease brains may indicate that if both are markers of reactive astrogliosis, they may detect different states or subtypes of astrogliosis. Increased 11C-DED brain binding seems to be an earlier phenomenon in Alzheimer's disease progression than increased plasma GFAP concentration.
Collapse
Affiliation(s)
- Konstantinos Chiotis
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Charlotte Johansson
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Elena Rodriguez-Vieitez
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| | - Caroline Graff
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
- Unit for Hereditary Dementia, Karolinska University Hospital-Solna, Solna, Sweden
| | - Agneta Nordberg
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden.
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
10
|
Fontana IC, Scarpa M, Malarte ML, Rocha FM, Ausellé-Bosch S, Bluma M, Bucci M, Chiotis K, Kumar A, Nordberg A. Astrocyte Signature in Alzheimer's Disease Continuum through a Multi-PET Tracer Imaging Perspective. Cells 2023; 12:1469. [PMID: 37296589 PMCID: PMC10253101 DOI: 10.3390/cells12111469] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/02/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
Reactive astrogliosis is an early event in the continuum of Alzheimer's disease (AD). Current advances in positron emission tomography (PET) imaging provide ways of assessing reactive astrogliosis in the living brain. In this review, we revisit clinical PET imaging and in vitro findings using the multi-tracer approach, and point out that reactive astrogliosis precedes the deposition of Aβ plaques, tau pathology, and neurodegeneration in AD. Furthermore, considering the current view of reactive astrogliosis heterogeneity-more than one subtype of astrocyte involved-in AD, we discuss how astrocytic body fluid biomarkers might fit into trajectories different from that of astrocytic PET imaging. Future research focusing on the development of innovative astrocytic PET radiotracers and fluid biomarkers may provide further insights into the heterogeneity of reactive astrogliosis and improve the detection of AD in its early stages.
Collapse
Affiliation(s)
- Igor C. Fontana
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Miriam Scarpa
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Mona-Lisa Malarte
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Filipa M. Rocha
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
- Instituto de Ciência Biomédicas Abel Salazar da Universidade do Porto, 4050-313 Porto, Portugal
- Faculdade de Engenharia, Universidade do Porto, 4200-465 Porto, Portugal
| | - Sira Ausellé-Bosch
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
- Faculty of Health and Life Sciences, Pompeu Fabra University, 08003 Barcelona, Spain
| | - Marina Bluma
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Marco Bucci
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Konstantinos Chiotis
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Amit Kumar
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, 141 57 Stockholm, Sweden
| |
Collapse
|
11
|
The Fuzzy Border between the Functional and Dysfunctional Effects of Beta-Amyloid: A Synaptocentric View of Neuron-Glia Entanglement. Biomedicines 2023; 11:biomedicines11020484. [PMID: 36831020 PMCID: PMC9953143 DOI: 10.3390/biomedicines11020484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/01/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Recent observations from clinical trials using monoclonal antibodies against Aβ seem to suggest that Aβ-targeting is modestly effective and not sufficiently based on an effective challenge of the role of Aβ from physiological to pathological. After an accelerated approval procedure for aducanumab, and more recently lecanemab, their efficacy and safety remain to be fully defined despite previous attempts with various monoclonal antibodies, and both academic institutions and pharmaceutical companies are actively searching for novel treatments. Aβ needs to be clarified further in a more complicated context, taking into account both its accumulation and its biological functions during the course of the disease. In this review, we discuss the border between activities affecting early, potentially reversible dysfunctions of the synapse and events trespassing the threshold of inflammatory, self-sustaining glial activation, leading to irreversible damage. We detail a clear understanding of the biological mechanisms underlying the derangement from function to dysfunction and the switch of the of Aβ role from physiological to pathological. A picture is emerging where the optimal therapeutic strategy against AD should involve a number of allied molecular processes, displaying efficacy not only in reducing the well-known AD pathogenesis players, such as Aβ or neuroinflammation, but also in preventing their adverse effects.
Collapse
|
12
|
Kumar A, Fontana IC, Nordberg A. Reactive astrogliosis: A friend or foe in the pathogenesis of Alzheimer's disease. J Neurochem 2023; 164:309-324. [PMID: 34931315 DOI: 10.1111/jnc.15565] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/28/2022]
Abstract
Astrocytes are highly efficient homeostatic glial cells playing a crucial role in optimal brain functioning and homeostasis. Astrocytes respond to changes in brain homoeostasis following central nervous system (CNS) injury/diseased state by a specific defence mechanism called reactive astrogliosis. Recent studies have implicated and placed reactive astrogliosis in the centre of pathophysiology of Alzheimer's disease (AD) and other neurodegenerative disorders. The AD biomarker field is evolving rapidly with new findings providing strong evidence which supports the notion that a reactive astrogliosis is an early event in the time course of AD progression which may precede other pathological hallmarks of AD. Clinical/translational in vivo PET and in vitro postmortem brain imaging studies demonstrated 'a first and second wave' of reactive astrogliosis in AD with distinct close-loop relationships with other pathological biomarkers at different stages of the disease. At the end stages, reactive astrocytes are found to be associated, or in proximity, with amyloid plaque and tau pathological deposits in postmortem AD brains. Several new PET-tracers, which are being in pipeline and validated at a very fast pace for mapping and visualising reactive astrogliosis in the brain, will provide further invaluable mechanistic insights into AD and other non-AD dementia pathologies. The complementary roles of microglia and astrocyte activation in AD progression, along with the clinical value of new fluid astrocytes biomarkers in the context of existing biomarkers, are the latest avenue that needs further exploration.
Collapse
Affiliation(s)
- Amit Kumar
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Igor C Fontana
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Theme Aging, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
13
|
Loftus JR, Puri S, Meyers SP. Multimodality imaging of neurodegenerative disorders with a focus on multiparametric magnetic resonance and molecular imaging. Insights Imaging 2023; 14:8. [PMID: 36645560 PMCID: PMC9842851 DOI: 10.1186/s13244-022-01358-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 12/13/2022] [Indexed: 01/17/2023] Open
Abstract
Neurodegenerative diseases afflict a large number of persons worldwide, with the prevalence and incidence of dementia rapidly increasing. Despite their prevalence, clinical diagnosis of dementia syndromes remains imperfect with limited specificity. Conventional structural-based imaging techniques also lack the accuracy necessary for confident diagnosis. Multiparametric magnetic resonance imaging and molecular imaging provide the promise of improving specificity and sensitivity in the diagnosis of neurodegenerative disease as well as therapeutic monitoring of monoclonal antibody therapy. This educational review will briefly focus on the epidemiology, clinical presentation, and pathologic findings of common and uncommon neurodegenerative diseases. Imaging features of each disease spanning from conventional magnetic resonance sequences to advanced multiparametric methods such as resting-state functional magnetic resonance imaging and arterial spin labeling imaging will be described in detail. Additionally, the review will explore the findings of each diagnosis on molecular imaging including single-photon emission computed tomography and positron emission tomography with a variety of clinically used and experimental radiotracers. The literature and clinical cases provided demonstrate the power of advanced magnetic resonance imaging and molecular techniques in the diagnosis of neurodegenerative diseases and areas of future and ongoing research. With the advent of combined positron emission tomography/magnetic resonance imaging scanners, hybrid protocols utilizing both techniques are an attractive option for improving the evaluation of neurodegenerative diseases.
Collapse
Affiliation(s)
- James Ryan Loftus
- grid.412750.50000 0004 1936 9166Department of Imaging Sciences, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14642 USA
| | - Savita Puri
- grid.412750.50000 0004 1936 9166Department of Imaging Sciences, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14642 USA
| | - Steven P. Meyers
- grid.412750.50000 0004 1936 9166Department of Imaging Sciences, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14642 USA
| |
Collapse
|
14
|
Kim J, Kim YK. Molecular Imaging of Neuroinflammation in Alzheimer's Disease and Mild Cognitive Impairment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:301-326. [PMID: 36949316 DOI: 10.1007/978-981-19-7376-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent neurocognitive disorder. Due to the ineffectiveness of treatments targeting the amyloid cascade, molecular biomarkers for neuroinflammation are attracting attention with increasing knowledge about the role of neuroinflammation in the pathogenesis of AD. This chapter will explore the results of studies using molecular imaging for diagnosing AD and mild cognitive impairment (MCI). Because it is critical to interpreting the data to understand which substances are targeted in molecular imaging, this chapter will discuss the two most significant targets, microglia and astrocytes, as well as the best-known radioligands for each. Then, neuroimaging results with PET neuroinflammation imaging will be reviewed for AD and MCI. Although a growing body of evidence has suggested that these molecular imaging biomarkers for neuroinflammation may have a role in the diagnosis of AD and MCI, the findings are inconsistent or cross-sectional, which indicates that it is difficult to apply the contents in practice due to the need for additional study. In particular, because the results of multiple interventions targeting neuroinflammation were inconclusive, molecular imaging markers for neuroinflammation can be used in combination with conventional markers to select appropriate patients for early intervention for neuroinflammation rather than as a single marker.
Collapse
Affiliation(s)
- Junhyung Kim
- Department of Psychiatry, Korea University College of Medicine, Korea University Guro Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Ku Kim
- Department of Psychiatry, Korea University Ansan Hospital, Ansan, Republic of Korea.
| |
Collapse
|
15
|
Huang Z, Lin HW(K, Zhang Q, Zong X. Targeting Alzheimer's Disease: The Critical Crosstalk between the Liver and Brain. Nutrients 2022; 14:nu14204298. [PMID: 36296980 PMCID: PMC9609624 DOI: 10.3390/nu14204298] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 01/24/2023] Open
Abstract
Alzheimer's disease (AD), an age-related neurodegenerative disorder, is currently incurable. Imbalanced amyloid-beta (Aβ) generation and clearance are thought to play a pivotal role in the pathogenesis of AD. Historically, strategies targeting Aβ clearance have typically focused on central clearance, but with limited clinical success. Recently, the contribution of peripheral systems, particularly the liver, to Aβ clearance has sparked an increased interest. In addition, AD presents pathological features similar to those of metabolic syndrome, and the critical involvement of brain energy metabolic disturbances in this disease has been recognized. More importantly, the liver may be a key regulator in these abnormalities, far beyond our past understanding. Here, we review recent animal and clinical findings indicating that liver dysfunction represents an early event in AD pathophysiology. We further propose that compromised peripheral Aβ clearance by the liver and aberrant hepatic physiological processes may contribute to AD neurodegeneration. The role of a hepatic synthesis product, fibroblast growth factor 21 (FGF21), in the management of AD is also discussed. A deeper understanding of the communication between the liver and brain may lead to new opportunities for the early diagnosis and treatment of AD.
Collapse
|
16
|
Huang J. Novel brain PET imaging agents: Strategies for imaging neuroinflammation in Alzheimer’s disease and mild cognitive impairment. Front Immunol 2022; 13:1010946. [PMID: 36211392 PMCID: PMC9537554 DOI: 10.3389/fimmu.2022.1010946] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/06/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disease with a concealed onset and continuous deterioration. Mild cognitive impairment (MCI) is the prodromal stage of AD. Molecule-based imaging with positron emission tomography (PET) is critical in tracking pathophysiological changes among AD and MCI patients. PET with novel targets is a promising approach for diagnostic imaging, particularly in AD patients. Our present review overviews the current status and applications of in vivo molecular imaging toward neuroinflammation. Although radiotracers can remarkably diagnose AD and MCI patients, a variety of limitations prevent the recommendation of a single technique. Recent studies examining neuroinflammation PET imaging suggest an alternative approach to evaluate disease progression. This review concludes that PET imaging towards neuroinflammation is considered a promising approach to deciphering the enigma of the pathophysiological process of AD and MCI.
Collapse
|
17
|
Zimmer ER, Pascoal TA, Rosa-Neto P, Nordberg A, Pellerin L. Comment on "Microglial activation states drive glucose uptake and FDG-PET alterations in neurodegenerative diseases". Sci Transl Med 2022; 14:eabm8302. [PMID: 36001681 DOI: 10.1126/scitranslmed.abm8302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Astrocytes might be the major contributor to the radioactive signal captured by PET in the microglia-dependent modulation of FDG-PET.
Collapse
Affiliation(s)
- Eduardo R Zimmer
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul (RS) 90035-003, Brazil
- Graduate Program in Biological Sciences: Biochemistry, UFRGS, Porto Alegre, RS 90035-003, Brazil
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics, UFRGS, Porto Alegre, RS 90035-003, Brazil
- Brain Institute of Rio Grande Do Sul, Pontifical Catholic University of Rio Grande Do Sul, Porto Alegre, RS 90610-000, Brazil
- McGill Centre for Studies in Aging, McGill University, Montreal, Quebec H4H 1R3, Canada
| | - Tharick A Pascoal
- McGill Centre for Studies in Aging, McGill University, Montreal, Quebec H4H 1R3, Canada
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, McGill University, Montreal, Quebec H4H 1R3, Canada
| | - Pedro Rosa-Neto
- McGill Centre for Studies in Aging, McGill University, Montreal, Quebec H4H 1R3, Canada
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, McGill University, Montreal, Quebec H4H 1R3, Canada
- Departments of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 1A1, Canada
- Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm 141 52, Sweden
- Theme Aging, Karolinska University Hospital, Stockholm 141 52, Sweden
| | - Luc Pellerin
- IRMETIST, Inserm U1313, University of Poitiers and CHU of Poitiers, Poitiers 86021, France
| |
Collapse
|
18
|
Andersen JV, Schousboe A, Verkhratsky A. Astrocyte energy and neurotransmitter metabolism in Alzheimer's disease: integration of the glutamate/GABA-glutamine cycle. Prog Neurobiol 2022; 217:102331. [PMID: 35872221 DOI: 10.1016/j.pneurobio.2022.102331] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 02/06/2023]
Abstract
Astrocytes contribute to the complex cellular pathology of Alzheimer's disease (AD). Neurons and astrocytes function in close collaboration through neurotransmitter recycling, collectively known as the glutamate/GABA-glutamine cycle, which is essential to sustain neurotransmission. Neurotransmitter recycling is intimately linked to astrocyte energy metabolism. In the course of AD, astrocytes undergo extensive metabolic remodeling, which may profoundly affect the glutamate/GABA-glutamine cycle. The consequences of altered astrocyte function and metabolism in relation to neurotransmitter recycling are yet to be comprehended. Metabolic alterations of astrocytes in AD deprive neurons of metabolic support, thereby contributing to synaptic dysfunction and neurodegeneration. In addition, several astrocyte-specific components of the glutamate/GABA-glutamine cycle, including glutamine synthesis and synaptic neurotransmitter uptake, are perturbed in AD. Integration of the complex astrocyte biology within the context of AD is essential for understanding the fundamental mechanisms of the disease, while restoring astrocyte metabolism may serve as an approach to arrest or even revert clinical progression of AD.
Collapse
Affiliation(s)
- Jens V Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; Achucarro Center for Neuroscience, IKERBASQUE, 48011 Bilbao, Spain; Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania.
| |
Collapse
|
19
|
Salvadó G, Milà-Alomà M, Shekari M, Ashton NJ, Operto G, Falcon C, Cacciaglia R, Minguillon C, Fauria K, Niñerola-Baizán A, Perissinotti A, Benedet AL, Kollmorgen G, Suridjan I, Wild N, Molinuevo JL, Zetterberg H, Blennow K, Suárez-Calvet M, Gispert JD. Reactive astrogliosis is associated with higher cerebral glucose consumption in the early Alzheimer's continuum. Eur J Nucl Med Mol Imaging 2022; 49:4567-4579. [PMID: 35849149 DOI: 10.1007/s00259-022-05897-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 06/28/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE Glial activation is one of the earliest mechanisms to be altered in Alzheimer's disease (AD). Glial fibrillary acidic protein (GFAP) relates to reactive astrogliosis and can be measured in both cerebrospinal fluid (CSF) and blood. Plasma GFAP has been suggested to become altered earlier in AD than its CSF counterpart. Although astrocytes consume approximately half of the glucose-derived energy in the brain, the relationship between reactive astrogliosis and cerebral glucose metabolism is poorly understood. Here, we aimed to investigate the association between fluorodeoxyglucose ([18F]FDG) uptake and reactive astrogliosis, by means of GFAP quantified in both plasma and CSF for the same participants. METHODS We included 314 cognitively unimpaired participants from the ALFA + cohort, 112 of whom were amyloid-β (Aβ) positive. Associations between GFAP markers and [18F]FDG uptake were studied. We also investigated whether these associations were modified by Aβ and tau status (AT stages). RESULTS Plasma GFAP was positively associated with glucose consumption in the whole brain, while CSF GFAP associations with [18F]FDG uptake were only observed in specific smaller areas like temporal pole and superior temporal lobe. These associations persisted when accounting for biomarkers of Aβ pathology but became negative in Aβ-positive and tau-positive participants (A + T +) in similar areas of AD-related hypometabolism. CONCLUSIONS Higher astrocytic reactivity, probably in response to early AD pathological changes, is related to higher glucose consumption. With the onset of tau pathology, the observed uncoupling between astrocytic biomarkers and glucose consumption might be indicative of a failure to sustain the higher energetic demands required by reactive astrocytes.
Collapse
Affiliation(s)
- Gemma Salvadó
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, C/ Wellington, 30, 08005, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Milà-Alomà
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, C/ Wellington, 30, 08005, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Mahnaz Shekari
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, C/ Wellington, 30, 08005, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Institute of Psychiatry, King's College London, Maurice Wohl Clinical Neuroscience Institute, Psychology & Neuroscience, London, UK.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Grégory Operto
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, C/ Wellington, 30, 08005, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Carles Falcon
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, C/ Wellington, 30, 08005, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Centro de Investigación Biomédica en Red Bioingeniería, (CIBER-BBN), Biomateriales Y Nanomedicina, Barcelona, Spain
| | - Raffaele Cacciaglia
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, C/ Wellington, 30, 08005, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Carolina Minguillon
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, C/ Wellington, 30, 08005, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Karine Fauria
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, C/ Wellington, 30, 08005, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Aida Niñerola-Baizán
- Centro de Investigación Biomédica en Red Bioingeniería, (CIBER-BBN), Biomateriales Y Nanomedicina, Barcelona, Spain.,Nuclear Medicine Department, Hospital Clínic Barcelona, Barcelona, Spain
| | - Andrés Perissinotti
- Centro de Investigación Biomédica en Red Bioingeniería, (CIBER-BBN), Biomateriales Y Nanomedicina, Barcelona, Spain.,Nuclear Medicine Department, Hospital Clínic Barcelona, Barcelona, Spain
| | - Andréa L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | | | | | | | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, C/ Wellington, 30, 08005, Barcelona, Spain.,H. Lundbeck A/S, Copenhagen, Denmark
| | - Henrik Zetterberg
- Universitat Pompeu Fabra, Barcelona, Spain.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK.,UK Dementia Research Institute at UCL, London, UK.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Universitat Pompeu Fabra, Barcelona, Spain.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, C/ Wellington, 30, 08005, Barcelona, Spain. .,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain. .,Servei de Neurologia, Hospital del Mar, Barcelona, Spain.
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, C/ Wellington, 30, 08005, Barcelona, Spain. .,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain. .,Centro de Investigación Biomédica en Red Bioingeniería, (CIBER-BBN), Biomateriales Y Nanomedicina, Barcelona, Spain.
| | | |
Collapse
|
20
|
Staging of Alzheimer's disease: past, present, and future perspectives. Trends Mol Med 2022; 28:726-741. [PMID: 35717526 DOI: 10.1016/j.molmed.2022.05.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 01/01/2023]
Abstract
For many years Alzheimer's disease (AD) was associated with the dementia stage of the disease, the tail end of a pathophysiological process that lasts approximately two decades. Whereas early disease staging assessments focused on progressive deterioration of clinical functioning, brain imaging with positron emission tomography (PET) and cerebrospinal fluid (CSF) biomarker studies highlighted the long preclinical phase of AD in which a cascade of detectable biological abnormalities precede cognitive decline. The recent proliferation of imaging and fluid biomarkers of AD pathophysiology provide an opportunity for the identification of several biological stages in the preclinical phase of AD. We discuss the use of clinical and biomarker information in past, present, and future staging of AD. We highlight potential applications of PET, CSF, and plasma biomarkers for staging AD severity in vivo.
Collapse
|
21
|
Gao L, Yang WY, Qi H, Sun CJ, Qin XM, Du GH. Unveiling the anti-senescence effects and senescence-associated secretory phenotype (SASP) inhibitory mechanisms of Scutellaria baicalensis Georgi in low glucose-induced astrocytes based on boolean network. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 99:153990. [PMID: 35202958 DOI: 10.1016/j.phymed.2022.153990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/31/2022] [Accepted: 02/10/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Astrocytes senescence has been demonstrated in the aging brain and Alzheimer's disease (AD). Moreover, lower glucose metabolism has been confirmed in the early stage of AD. However, whether low glucose could induce astrocytes senescence remain ambiguous. Studies have shown that the ethanol extracts of Scutellaria baicalensis Georgi (SGE) exert neuroprotective and anti-aging effects, while whether SGE could delay astrocytes senescence was unclear. PURPOSE This study investigated the anti-senescence effect of SGE in low glucose-induced T98G cells and primary astrocytes, and explored the possible mechanisms based on boolean network. METHODS The neuroprotective effects of SGE in low glucose-induced T98G cells were evaluated by measurement of cell viability, LDH, ROS and ATP. The anti-senescence effects of SGE were investigated by detection of senescence-associated β-galactosidase (SA-β-Gal), senescence-associated secretory phenotype (SASP), cell cycle and senescence-related markers. The possible mechanisms of SGE in delaying astrocytes senescence were discovered through integrating transcriptomics with boolean network, and validation experiments were further performed. RESULTS Our results revealed that low glucose could induce astrocytes senescence, and SGE could delay astrocytes senescence by decreasing the staining rate of SA-β-gal, reducing secretions of SASP factors (IL-6, CXCL1, MMP-1), alleviating cell cycle arrest in G0/G1 phase, decreasing the formation of punctate DNA foci and down-regulating the expression of p16INK4A, p21 and γH2A.X. Transcriptomics and further verification results showed that SGE could markedly inhibit the mRNA expression levels of SASP factors (CXCL10, CXCL2, CCL2, IL-6, CXCR4, CCR7). Moreover, C-X-C motif chemokine 10 (CXCL10) was predicted to be the key SASP factor affecting the network stability by using boolean network. Further experiments validated that SGE could markedly reduce CXCL10 level, decrease the secretion of IL-6 and inhibit cell migration in CXCL10 induced primary astrocytes. CONCLUSION In summary, our research unmasks that the anti-senescence effects of SGE were highly correlated with the suppression of SASP secretions, and CXCL10 mediated the SASP inhibition effect of SGE in low glucose-induced astrocytes. Our study highlights that the delay of astrocytes senescence and the inhibition of SASP might be a new mechanism of SGE for alleviating neurodegenerative diseases such as AD.
Collapse
Affiliation(s)
- Li Gao
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, China.
| | - Wu-Yan Yang
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, China
| | - Hong Qi
- Complex Systems Research Center, Shanxi University, Taiyuan, China
| | - Chang-Jun Sun
- Complex Systems Research Center, Shanxi University, Taiyuan, China
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, China
| | - Guan-Hua Du
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
22
|
Garland EF, Hartnell IJ, Boche D. Microglia and Astrocyte Function and Communication: What Do We Know in Humans? Front Neurosci 2022; 16:824888. [PMID: 35250459 PMCID: PMC8888691 DOI: 10.3389/fnins.2022.824888] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/24/2022] [Indexed: 12/11/2022] Open
Abstract
Microglia and astrocytes play essential roles in the central nervous system contributing to many functions including homeostasis, immune response, blood-brain barrier maintenance and synaptic support. Evidence has emerged from experimental models of glial communication that microglia and astrocytes influence and coordinate each other and their effects on the brain environment. However, due to the difference in glial cells between humans and rodents, it is essential to confirm the relevance of these findings in human brains. Here, we aim to review the current knowledge on microglia-astrocyte crosstalk in humans, exploring novel methodological techniques used in health and disease conditions. This will include an in-depth look at cell culture and iPSCs, post-mortem studies, imaging and fluid biomarkers, genetics and transcriptomic data. In this review, we will discuss the advantages and limitations of these methods, highlighting the understanding these methods have brought the field on these cells communicative abilities, and the knowledge gaps that remain.
Collapse
Affiliation(s)
| | | | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
23
|
Harada R, Furumoto S, Kudo Y, Yanai K, Villemagne VL, Okamura N. Imaging of Reactive Astrogliosis by Positron Emission Tomography. Front Neurosci 2022; 16:807435. [PMID: 35210989 PMCID: PMC8862631 DOI: 10.3389/fnins.2022.807435] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/14/2022] [Indexed: 11/13/2022] Open
Abstract
Many neurodegenerative diseases are neuropathologically characterized by neuronal loss, gliosis, and the deposition of misfolded proteins such as β-amyloid (Aβ) plaques and tau tangles in Alzheimer’s disease (AD). In postmortem AD brains, reactive astrocytes and activated microglia are observed surrounding Aβ plaques and tau tangles. These activated glial cells secrete pro-inflammatory cytokines and reactive oxygen species, which may contribute to neurodegeneration. Therefore, in vivo imaging of glial response by positron emission tomography (PET) combined with Aβ and tau PET would provide new insights to better understand the disease process, as well as aid in the differential diagnosis, and monitoring glial response disease-specific therapeutics. There are two promising targets proposed for imaging reactive astrogliosis: monoamine oxidase-B (MAO-B) and imidazoline2 binding site (I2BS), which are predominantly expressed in the mitochondrial membranes of astrocytes and are upregulated in various neurodegenerative conditions. PET tracers targeting these two MAO-B and I2BS have been evaluated in humans. [18F]THK-5351, which was originally designed to target tau aggregates in AD, showed high affinity for MAO-B and clearly visualized reactive astrocytes in progressive supranuclear palsy (PSP). However, the lack of selectivity of [18F]THK-5351 binding to both MAO-B and tau, severely limits its clinical utility as a biomarker. Recently, [18F]SMBT-1 was developed as a selective and reversible MAO-B PET tracer via compound optimization of [18F]THK-5351. In this review, we summarize the strategy underlying molecular imaging of reactive astrogliosis and clinical studies using MAO-B and I2BS PET tracers.
Collapse
Affiliation(s)
- Ryuichi Harada
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
- *Correspondence: Ryuichi Harada,
| | - Shozo Furumoto
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Yukitsuka Kudo
- Department of New Therapeutics Innovation for Alzheimer’s and Dementia, Institute of Development and Aging, Tohoku University, Sendai, Japan
| | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Victor L. Villemagne
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
- Nobuyuki Okamura,
| |
Collapse
|
24
|
Beard E, Lengacher S, Dias S, Magistretti PJ, Finsterwald C. Astrocytes as Key Regulators of Brain Energy Metabolism: New Therapeutic Perspectives. Front Physiol 2022; 12:825816. [PMID: 35087428 PMCID: PMC8787066 DOI: 10.3389/fphys.2021.825816] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/20/2021] [Indexed: 12/11/2022] Open
Abstract
Astrocytes play key roles in the regulation of brain energy metabolism, which has a major impact on brain functions, including memory, neuroprotection, resistance to oxidative stress and homeostatic tone. Energy demands of the brain are very large, as they continuously account for 20–25% of the whole body’s energy consumption. Energy supply of the brain is tightly linked to neuronal activity, providing the origin of the signals detected by the widely used functional brain imaging techniques such as functional magnetic resonance imaging and positron emission tomography. In particular, neuroenergetic coupling is regulated by astrocytes through glutamate uptake that triggers astrocytic aerobic glycolysis and leads to glucose uptake and lactate release, a mechanism known as the Astrocyte Neuron Lactate Shuttle. Other neurotransmitters such as noradrenaline and Vasoactive Intestinal Peptide mobilize glycogen, the reserve for glucose exclusively localized in astrocytes, also resulting in lactate release. Lactate is then transferred to neurons where it is used, after conversion to pyruvate, as a rapid energy substrate, and also as a signal that modulates neuronal excitability, homeostasis, and the expression of survival and plasticity genes. Importantly, glycolysis in astrocytes and more generally cerebral glucose metabolism progressively deteriorate in aging and age-associated neurodegenerative diseases such as Alzheimer’s disease. This decreased glycolysis actually represents a common feature of several neurological pathologies. Here, we review the critical role of astrocytes in the regulation of brain energy metabolism, and how dysregulation of astrocyte-mediated metabolic pathways is involved in brain hypometabolism. Further, we summarize recent efforts at preclinical and clinical stages to target brain hypometabolism for the development of new therapeutic interventions in age-related neurodegenerative diseases.
Collapse
|
25
|
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: 3] [Impact Index Per Article: 1.0] [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
|
26
|
Zhou R, Ji B, Kong Y, Qin L, Ren W, Guan Y, Ni R. PET Imaging of Neuroinflammation in Alzheimer's Disease. Front Immunol 2021; 12:739130. [PMID: 34603323 PMCID: PMC8481830 DOI: 10.3389/fimmu.2021.739130] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 08/27/2021] [Indexed: 12/15/2022] Open
Abstract
Neuroinflammation play an important role in Alzheimer's disease pathogenesis. Advances in molecular imaging using positron emission tomography have provided insights into the time course of neuroinflammation and its relation with Alzheimer's disease central pathologies in patients and in animal disease models. Recent single-cell sequencing and transcriptomics indicate dynamic disease-associated microglia and astrocyte profiles in Alzheimer's disease. Mitochondrial 18-kDa translocator protein is the most widely investigated target for neuroinflammation imaging. New generation of translocator protein tracers with improved performance have been developed and evaluated along with tau and amyloid imaging for assessing the disease progression in Alzheimer's disease continuum. Given that translocator protein is not exclusively expressed in glia, alternative targets are under rapid development, such as monoamine oxidase B, matrix metalloproteinases, colony-stimulating factor 1 receptor, imidazoline-2 binding sites, cyclooxygenase, cannabinoid-2 receptor, purinergic P2X7 receptor, P2Y12 receptor, the fractalkine receptor, triggering receptor expressed on myeloid cells 2, and receptor for advanced glycation end products. Promising targets should demonstrate a higher specificity for cellular locations with exclusive expression in microglia or astrocyte and activation status (pro- or anti-inflammatory) with highly specific ligand to enable in vivo brain imaging. In this review, we summarised recent advances in the development of neuroinflammation imaging tracers and provided an outlook for promising targets in the future.
Collapse
Affiliation(s)
- Rong Zhou
- Department of Nephrology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bin Ji
- Department of Radiopharmacy and Molecular Imaging, School of Pharmacy, Fudan University, Shanghai, China
| | - Yanyan Kong
- Positron Emission Tomography (PET) Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Limei Qin
- Inner Mongolia Baicaotang Qin Chinese Mongolia Hospital, Hohhot, China
| | - Wuwei Ren
- School of Information Science and Technology, Shanghaitech University, Shanghai, China
| | - Yihui Guan
- Positron Emission Tomography (PET) Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, University of Zurich & Eidgenössische Technische Hochschule Zürich (ETH Zurich), Zurich, Switzerland
| |
Collapse
|
27
|
Bellaver B, Ferrari-Souza JP, Uglione da Ros L, Carter SF, Rodriguez-Vieitez E, Nordberg A, Pellerin L, Rosa-Neto P, Leffa DT, Zimmer ER. Astrocyte Biomarkers in Alzheimer Disease: A Systematic Review and Meta-analysis. Neurology 2021; 96:e2944-e2955. [PMID: 33952650 DOI: 10.1212/wnl.0000000000012109] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/19/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To perform a systematic review and meta-analysis to determine whether fluid and imaging astrocyte biomarkers are altered in Alzheimer disease (AD). METHODS PubMed and Web of Science databases were searched for articles reporting fluid or imaging astrocyte biomarkers in AD. Pooled effect sizes were determined with standardized mean differences (SMDs) using the Hedge G method with random effects to determine biomarker performance. Adapted questions from the Quality Assessment of Diagnostic Accuracy Studies were applied for quality assessment. A protocol for this study has been previously registered in PROSPERO (registration number: CRD42020192304). RESULTS The initial search identified 1,425 articles. After exclusion criteria were applied, 33 articles (a total of 3,204 individuals) measuring levels of glial fibrillary acidic protein (GFAP), S100B, chitinase-3-like protein 1 (YKL-40), and aquaporin 4 in the blood and CSF, as well as monoamine oxidase-B indexed by PET 11C-deuterium-l-deprenyl, were included. GFAP (SMD 0.94, 95% confidence interval [CI] 0.71-1.18) and YKL-40 (SMD 0.76, 95% CI 0.63-0.89) levels in the CSF and S100B levels in the blood (SMD 2.91, 95% CI 1.01-4.8) were found to be significantly increased in patients with AD. CONCLUSIONS Despite significant progress, applications of astrocyte biomarkers in AD remain in their early days. This meta-analysis demonstrated that astrocyte biomarkers are consistently altered in AD and supports further investigation for their inclusion in the AD clinical research framework for observational and interventional studies.
Collapse
Affiliation(s)
- Bruna Bellaver
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - João Pedro Ferrari-Souza
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Lucas Uglione da Ros
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Stephen F Carter
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Elena Rodriguez-Vieitez
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Agneta Nordberg
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Luc Pellerin
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Pedro Rosa-Neto
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Douglas Teixeira Leffa
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Eduardo R Zimmer
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil.
| |
Collapse
|
28
|
Chaney AM, Lopez-Picon FR, Serrière S, Wang R, Bochicchio D, Webb SD, Vandesquille M, Harte MK, Georgiadou C, Lawrence C, Busson J, Vercouillie J, Tauber C, Buron F, Routier S, Reekie T, Snellman A, Kassiou M, Rokka J, Davies KE, Rinne JO, Salih DA, Edwards FA, Orton LD, Williams SR, Chalon S, Boutin H. Prodromal neuroinflammatory, cholinergic and metabolite dysfunction detected by PET and MRS in the TgF344-AD transgenic rat model of AD: a collaborative multi-modal study. Am J Cancer Res 2021; 11:6644-6667. [PMID: 34093845 PMCID: PMC8171096 DOI: 10.7150/thno.56059] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/15/2021] [Indexed: 12/25/2022] Open
Abstract
Mouse models of Alzheimer's disease (AD) are valuable but do not fully recapitulate human AD pathology, such as spontaneous Tau fibril accumulation and neuronal loss, necessitating the development of new AD models. The transgenic (TG) TgF344-AD rat has been reported to develop age-dependent AD features including neuronal loss and neurofibrillary tangles, despite only expressing APP and PSEN1 mutations, suggesting an improved modelling of AD hallmarks. Alterations in neuronal networks as well as learning performance and cognition tasks have been reported in this model, but none have combined a longitudinal, multimodal approach across multiple centres, which mimics the approaches commonly taken in clinical studies. We therefore aimed to further characterise the progression of AD-like pathology and cognition in the TgF344-AD rat from young-adults (6 months (m)) to mid- (12 m) and advanced-stage (18 m, 25 m) of the disease. Methods: TgF344-AD rats and wild-type (WT) littermates were imaged at 6 m, 12 m and 18 m with [18F]DPA-714 (TSPO, neuroinflammation), [18F]Florbetaben (Aβ) and [18F]ASEM (α7-nicotinic acetylcholine receptor) and with magnetic resonance spectroscopy (MRS) and with (S)-[18F]THK5117 (Tau) at 15 and 25 m. Behaviour tests were also performed at 6 m, 12 m and 18 m. Immunohistochemistry (CD11b, GFAP, Aβ, NeuN, NeuroChrom) and Tau (S)-[18F]THK5117 autoradiography, immunohistochemistry and Western blot were also performed. Results: [18F]DPA-714 positron emission tomography (PET) showed an increase in neuroinflammation in TG vs wildtype animals from 12 m in the hippocampus (+11%), and at the advanced-stage AD in the hippocampus (+12%), the thalamus (+11%) and frontal cortex (+14%). This finding coincided with strong increases in brain microgliosis (CD11b) and astrogliosis (GFAP) at these time-points as assessed by immunohistochemistry. In vivo [18F]ASEM PET revealed an age-dependent increase uptake in the striatum and pallidum/nucleus basalis of Meynert in WT only, similar to that observed with this tracer in humans, resulting in TG being significantly lower than WT by 18 m. In vivo [18F]Florbetaben PET scanning detected Aβ accumulation at 18 m, and (S)-[18F]THK5117 PET revealed subsequent Tau accumulation at 25m in hippocampal and cortical regions. Aβ plaques were low but detectable by immunohistochemistry from 6 m, increasing further at 12 and 18 m with Tau-positive neurons adjacent to Aβ plaques at 18 m. NeuroChrom (a pan neuronal marker) immunohistochemistry revealed a loss of neuronal staining at the Aβ plaques locations, while NeuN labelling revealed an age-dependent decrease in hippocampal neuron number in both genotypes. Behavioural assessment using the novel object recognition task revealed that both WT & TgF344-AD animals discriminated the novel from familiar object at 3 m and 6 m of age. However, low levels of exploration observed in both genotypes at later time-points resulted in neither genotype successfully completing the task. Deficits in social interaction were only observed at 3 m in the TgF344-AD animals. By in vivo MRS, we showed a decrease in neuronal marker N-acetyl-aspartate in the hippocampus at 18 m (-18% vs age-matched WT, and -31% vs 6 m TG) and increased Taurine in the cortex of TG (+35% vs age-matched WT, and +55% vs 6 m TG). Conclusions: This multi-centre multi-modal study demonstrates, for the first time, alterations in brain metabolites, cholinergic receptors and neuroinflammation in vivo in this model, validated by robust ex vivo approaches. Our data confirm that, unlike mouse models, the TgF344-AD express Tau pathology that can be detected via PET, albeit later than by ex vivo techniques, and is a useful model to assess and longitudinally monitor early neurotransmission dysfunction and neuroinflammation in AD.
Collapse
|
29
|
Ni R, Röjdner J, Voytenko L, Dyrks T, Thiele A, Marutle A, Nordberg A. In vitro Characterization of the Regional Binding Distribution of Amyloid PET Tracer Florbetaben and the Glia Tracers Deprenyl and PK11195 in Autopsy Alzheimer's Brain Tissue. J Alzheimers Dis 2021; 80:1723-1737. [PMID: 33749648 PMCID: PMC8150513 DOI: 10.3233/jad-201344] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Emerging evidence indicates a central role of gliosis in Alzheimer's disease (AD) pathophysiology. However, the regional distribution and interaction of astrogliosis and microgliosis in association with amyloid-β (Aβ) still remain uncertain. OBJECTIVE Here we studied the pathological profiles in autopsy AD brain by using specific imaging tracers. METHODS Autopsy brain tissues of AD (n = 15, age 70.4±8.5 years) and control cases (n = 12, age 76.6±10.9) were examined with homogenate binding assays, autoradiography for Aβ plaques (3H-florbetaben/3H-PIB), astrogliosis (3H-L-deprenyl), and microgliosis (3H-PK11195/3H-FEMPA), as well as immunoassays. RESULTS In vitro saturation analysis revealed high-affinity binding sites of 3H-florbetaben, 3H-L-deprenyl, and 3H-PK11195/3H-FEMPA in the frontal cortex of AD cases. In vitro3H-florbetaben binding increased across cortical and subcortical regions of AD compared to control with the highest binding in the frontal and parietal cortices. The in vitro3H-L-deprenyl binding showed highest binding in the hippocampus (dentate gyrus) followed by cortical and subcortical regions of AD while the GFAP expression was upregulated only in the hippocampus compared to control. The in vitro3H-PK11195 binding was solely increased in the parietal cortex and the hippocampus of AD compared to control. The 3H-florbetaben binding positively correlated with the 3H-L-deprenyl binding in the hippocampus and parietal cortex of AD and controls. Similarly, a positive correlation was observed between 3H-florbetaben binding and GFAP expression in hippocampus of AD and control. CONCLUSION The use of multi-imaging tracers revealed different regional pattern of changes in autopsy AD brain with respect to amyloid plaque pathology versus astrogliosis and microgliosis.
Collapse
Affiliation(s)
- Ruiqing Ni
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Jennie Röjdner
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Larysa Voytenko
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Amelia Marutle
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Theme Aging, The Aging Brain Unit, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
30
|
Narayanaswami V, Tong J, Schifani C, Bloomfield PM, Dahl K, Vasdev N. Preclinical Evaluation of TSPO and MAO-B PET Radiotracers in an LPS Model of Neuroinflammation. PET Clin 2021; 16:233-247. [PMID: 33648665 DOI: 10.1016/j.cpet.2020.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Discovery of novel PET radiotracers targeting neuroinflammation (microglia and astrocytes) is actively pursued. Employing a lipopolysaccharide (LPS) rat model, this longitudinal study evaluated the translocator protein 18-kDa radiotracer [18F]FEPPA (primarily microglia) and monoamine oxidase B radiotracers [11C]L-deprenyl and [11C]SL25.1188 (astrocytes preferred). Increased [18F]FEPPA binding peaked at 1 week in LPS-injected striatum whereas increased lazabemide-sensitive [11C]L-deprenyl binding developed later. No increase in radiotracer uptake was observed for [11C]SL25.1188. The unilateral intrastriatal LPS rat model may serve as a useful tool for benchmarking PET tracers targeted toward distinct phases of neuroinflammatory reactions involving both microglia and astrocytes.
Collapse
Affiliation(s)
- Vidya Narayanaswami
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Room 270, Toronto, Ontario M5T 1R8, Canada
| | - Junchao Tong
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Room 339, Toronto, Ontario M5T 1R8, Canada
| | - Christin Schifani
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Room 270, Toronto, Ontario M5T 1R8, Canada
| | - Peter M Bloomfield
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Room B26A, Toronto, Ontario M5T 1R8, Canada
| | - Kenneth Dahl
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Room B02, Toronto, Ontario M5T 1R8, Canada
| | - Neil Vasdev
- Department of Psychiatry, Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, Centre for Addiction and Mental Health, University of Toronto, 250 College Street, Room PET G2, Toronto, Ontario M5T 1R8, Canada.
| |
Collapse
|
31
|
Villemagne VL, Barkhof F, Garibotto V, Landau SM, Nordberg A, van Berckel BNM. Molecular Imaging Approaches in Dementia. Radiology 2021; 298:517-530. [PMID: 33464184 DOI: 10.1148/radiol.2020200028] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The increasing prevalence of dementia worldwide places a high demand on healthcare providers to perform a diagnostic work-up in relatively early stages of the disease, given that the pathologic process usually begins decades before symptoms are evident. Structural imaging is recommended to rule out other disorders and can only provide diagnosis in a late stage with limited specificity. Where PET imaging previously focused on the spatial pattern of hypometabolism, the past decade has seen the development of novel tracers to demonstrate characteristic protein abnormalities. Molecular imaging using PET/SPECT is able to show amyloid and tau deposition in Alzheimer disease and dopamine depletion in parkinsonian disorders starting decades before symptom onset. Novel tracers for neuroinflammation and synaptic density are being developed to further unravel the molecular pathologic characteristics of dementia disorders. In this article, the authors review the current status of established and emerging PET tracers in a diagnostic setting and also their value as prognostic markers in research studies and outcome measures for clinical trials in Alzheimer disease.
Collapse
Affiliation(s)
- Victor L Villemagne
- From the Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pa (V.L.V.); Department of Medicine, the University of Melbourne, Melbourne, Australia (V.L.V.); Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, the Netherlands (F.B., B.N.M.v.B.); UCL institutes of Neurology and Healthcare Engineering, London, England (F.B.); Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals and Laboratory of Neuroimaging and Innovative Molecular Tracers, Geneva University, Geneva, Switzerland (V.G.); Helen Wills Neuroscience Institute, University of California, Berkeley, Calif (S.M.L.); Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, Calif (S.M.L.); Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden (A.N.); and Theme Aging, Karolinska University Hospital, Stockholm, Sweden (A.N.)
| | - Frederik Barkhof
- From the Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pa (V.L.V.); Department of Medicine, the University of Melbourne, Melbourne, Australia (V.L.V.); Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, the Netherlands (F.B., B.N.M.v.B.); UCL institutes of Neurology and Healthcare Engineering, London, England (F.B.); Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals and Laboratory of Neuroimaging and Innovative Molecular Tracers, Geneva University, Geneva, Switzerland (V.G.); Helen Wills Neuroscience Institute, University of California, Berkeley, Calif (S.M.L.); Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, Calif (S.M.L.); Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden (A.N.); and Theme Aging, Karolinska University Hospital, Stockholm, Sweden (A.N.)
| | - Valentina Garibotto
- From the Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pa (V.L.V.); Department of Medicine, the University of Melbourne, Melbourne, Australia (V.L.V.); Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, the Netherlands (F.B., B.N.M.v.B.); UCL institutes of Neurology and Healthcare Engineering, London, England (F.B.); Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals and Laboratory of Neuroimaging and Innovative Molecular Tracers, Geneva University, Geneva, Switzerland (V.G.); Helen Wills Neuroscience Institute, University of California, Berkeley, Calif (S.M.L.); Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, Calif (S.M.L.); Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden (A.N.); and Theme Aging, Karolinska University Hospital, Stockholm, Sweden (A.N.)
| | - Susan M Landau
- From the Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pa (V.L.V.); Department of Medicine, the University of Melbourne, Melbourne, Australia (V.L.V.); Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, the Netherlands (F.B., B.N.M.v.B.); UCL institutes of Neurology and Healthcare Engineering, London, England (F.B.); Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals and Laboratory of Neuroimaging and Innovative Molecular Tracers, Geneva University, Geneva, Switzerland (V.G.); Helen Wills Neuroscience Institute, University of California, Berkeley, Calif (S.M.L.); Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, Calif (S.M.L.); Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden (A.N.); and Theme Aging, Karolinska University Hospital, Stockholm, Sweden (A.N.)
| | - Agneta Nordberg
- From the Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pa (V.L.V.); Department of Medicine, the University of Melbourne, Melbourne, Australia (V.L.V.); Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, the Netherlands (F.B., B.N.M.v.B.); UCL institutes of Neurology and Healthcare Engineering, London, England (F.B.); Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals and Laboratory of Neuroimaging and Innovative Molecular Tracers, Geneva University, Geneva, Switzerland (V.G.); Helen Wills Neuroscience Institute, University of California, Berkeley, Calif (S.M.L.); Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, Calif (S.M.L.); Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden (A.N.); and Theme Aging, Karolinska University Hospital, Stockholm, Sweden (A.N.)
| | - Bart N M van Berckel
- From the Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pa (V.L.V.); Department of Medicine, the University of Melbourne, Melbourne, Australia (V.L.V.); Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, the Netherlands (F.B., B.N.M.v.B.); UCL institutes of Neurology and Healthcare Engineering, London, England (F.B.); Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals and Laboratory of Neuroimaging and Innovative Molecular Tracers, Geneva University, Geneva, Switzerland (V.G.); Helen Wills Neuroscience Institute, University of California, Berkeley, Calif (S.M.L.); Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, Calif (S.M.L.); Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden (A.N.); and Theme Aging, Karolinska University Hospital, Stockholm, Sweden (A.N.)
| |
Collapse
|
32
|
Astroglial tracer BU99008 detects multiple binding sites in Alzheimer's disease brain. Mol Psychiatry 2021; 26:5833-5847. [PMID: 33888872 PMCID: PMC8758481 DOI: 10.1038/s41380-021-01101-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/17/2021] [Accepted: 04/01/2021] [Indexed: 12/15/2022]
Abstract
With reactive astrogliosis being established as one of the hallmarks of Alzheimer's disease (AD), there is high interest in developing novel positron emission tomography (PET) tracers to detect early astrocyte reactivity. BU99008, a novel astrocytic PET ligand targeting imidazoline-2 binding sites (I2BS) on astrocytes, might be a suitable candidate. Here we demonstrate for the first time that BU99008 could visualise reactive astrogliosis in postmortem AD brains and propose a multiple binding site [Super-high-affinity (SH), High-affinity (HA) and Low-affinity (LA)] model for BU99008, I2BS specific ligands (2-BFI and BU224) and deprenyl in AD and control (CN) brains. The proportion (%) and affinities of these sites varied significantly between the BU99008, 2-BFI, BU224 and deprenyl in AD and CN brains. Regional binding studies demonstrated significantly higher 3H-BU99008 binding in AD brain regions compared to CN. Comparative autoradiography studies reinforced these findings, showing higher specific binding for 3H-BU99008 than 3H-Deprenyl in sporadic AD brain compared to CN, implying that they might have different targets. The data clearly shows that BU99008 could detect I2BS expressing reactive astrocytes with good selectivity and specificity and hence be a potential attractive clinical astrocytic PET tracer for gaining further insight into the role of reactive astrogliosis in AD.
Collapse
|
33
|
Hendrix RD, Ou Y, Davis JE, Odle AK, Groves TR, Allen AR, Childs GV, Barger SW. Alzheimer amyloid-β- peptide disrupts membrane localization of glucose transporter 1 in astrocytes: implications for glucose levels in brain and blood. Neurobiol Aging 2020; 97:73-88. [PMID: 33161213 PMCID: PMC7736209 DOI: 10.1016/j.neurobiolaging.2020.10.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 08/25/2020] [Accepted: 10/02/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer’s disease (AD) is associated with disturbances in blood glucose regulation, and type-2 diabetes elevates the risk for dementia. A role for amyloid-β peptide (Aβ) in linking these age-related conditions has been proposed, tested primarily in transgenic mouse lines that overexpress mutated amyloid precursor protein (APP). Because APP has its own impacts on glucose regulation, we examined the BRI-Aβ42 line (“Aβ42-tg”), which produces extracellular Aβ1–42 in the CNS without elevation of APP. We also looked for interactions with diet-induced obesity (DIO) resulting from a high-fat, high-sucrose (“western”) diet. Aβ42-tg mice were impaired in both spatial memory and glucose tolerance. Although DIO induced insulin resistance, Aβ1–42 accumulation did not, and the impacts of DIO and Aβ on glucose tolerance were merely additive. Aβ42-tg mice exhibited no significant differences from wild-type in insulin production, body weight, lipidemia, appetite, physical activity, respiratory quotient, an-/orexigenic factors, or inflammatory factors. These negative findings suggested that the phenotype in these mice arose from perturbation of glucose excursion in an insulin-independent tissue. To wit, cerebral cortex of Aβ42-tg mice had reduced glucose utilization, similar to human patients with AD. This was associated with insufficient trafficking of glucose transporter 1 to the plasma membrane in parenchymal brain cells, a finding also documented in human AD tissue. Together, the lower cerebral metabolic rate of glucose and diminished function of parenchymal glucose transporter 1 indicate that aberrant regulation of blood glucose in AD likely reflects a central phenomenon, resulting from the effects of Aβ on cerebral parenchyma, rather than a generalized disruption of hypothalamic or peripheral endocrinology. The involvement of a specific glucose transporter in this deficit provides a new target for the design of AD therapies.
Collapse
Affiliation(s)
- Rachel D Hendrix
- Department of Neurobiology & Developmental Sciences, Little Rock, AR, USA
| | - Yang Ou
- Department of Geriatrics, Little Rock, AR, USA
| | - Jakeira E Davis
- Graduate Program in Interdisciplinary Biomedical Sciences, Little Rock, AR, USA
| | - Angela K Odle
- Department of Neurobiology & Developmental Sciences, Little Rock, AR, USA
| | - Thomas R Groves
- Department of Neurobiology & Developmental Sciences, Little Rock, AR, USA
| | - Antiño R Allen
- Department of Neurobiology & Developmental Sciences, Little Rock, AR, USA; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Gwen V Childs
- Department of Neurobiology & Developmental Sciences, Little Rock, AR, USA
| | - Steven W Barger
- Department of Neurobiology & Developmental Sciences, Little Rock, AR, USA; Department of Geriatrics, Little Rock, AR, USA; Geriatric Research, Education & Clinical Center, Central Arkansas Veterans Healthcare System, Little Rock, AR, USA.
| |
Collapse
|
34
|
Koller EJ, Chakrabarty P. Tau-Mediated Dysregulation of Neuroplasticity and Glial Plasticity. Front Mol Neurosci 2020; 13:151. [PMID: 32973446 PMCID: PMC7472665 DOI: 10.3389/fnmol.2020.00151] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/20/2020] [Indexed: 01/14/2023] Open
Abstract
The inability of individual neurons to compensate for aging-related damage leads to a gradual loss of functional plasticity in the brain accompanied by progressive impairment in learning and memory. Whereas this loss in neuroplasticity is gradual during normal aging, in neurodegenerative diseases such as Alzheimer’s disease (AD), this loss is accelerated dramatically, leading to the incapacitation of patients within a decade of onset of cognitive symptoms. The mechanisms that underlie this accelerated loss of neuroplasticity in AD are still not completely understood. While the progressively increasing proteinopathy burden, such as amyloid β (Aβ) plaques and tau tangles, definitely contribute directly to a neuron’s functional demise, the role of non-neuronal cells in controlling neuroplasticity is slowly being recognized as another major factor. These non-neuronal cells include astrocytes, microglia, and oligodendrocytes, which through regulating brain homeostasis, structural stability, and trophic support, play a key role in maintaining normal functioning and resilience of the neuronal network. It is believed that chronic signaling from these cells affects the homeostatic network of neuronal and non-neuronal cells to an extent to destabilize this harmonious milieu in neurodegenerative diseases like AD. Here, we will examine the experimental evidence regarding the direct and indirect pathways through which astrocytes and microglia can alter brain plasticity in AD, specifically as they relate to the development and progression of tauopathy. In this review article, we describe the concepts of neuroplasticity and glial plasticity in healthy aging, delineate possible mechanisms underlying tau-induced plasticity dysfunction, and discuss current clinical trials as well as future disease-modifying approaches.
Collapse
Affiliation(s)
- Emily J Koller
- Department of Neuroscience, University of Florida, Gainesville, FL, United States.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, United States
| | - Paramita Chakrabarty
- Department of Neuroscience, University of Florida, Gainesville, FL, United States.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, United States.,McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
35
|
Cavaliere C, Tramontano L, Fiorenza D, Alfano V, Aiello M, Salvatore M. Gliosis and Neurodegenerative Diseases: The Role of PET and MR Imaging. Front Cell Neurosci 2020; 14:75. [PMID: 32327973 PMCID: PMC7161920 DOI: 10.3389/fncel.2020.00075] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 03/13/2020] [Indexed: 12/16/2022] Open
Abstract
Glial activation characterizes most neurodegenerative and psychiatric diseases, often anticipating clinical manifestations and macroscopical brain alterations. Although imaging techniques have improved diagnostic accuracy in many neurological conditions, often supporting diagnosis, prognosis prediction and treatment outcome, very few molecular imaging probes, specifically focused on microglial and astrocytic activation, have been translated to a clinical setting. In this context, hybrid positron emission tomography (PET)/magnetic resonance (MR) scanners represent the most advanced tool for molecular imaging, combining the functional specificity of PET radiotracers (e.g., targeting metabolism, hypoxia, and inflammation) to both high-resolution and multiparametric information derived by MR in a single imaging acquisition session. This simultaneity of findings achievable by PET/MR, if useful for reciprocal technical adjustments regarding temporal and spatial cross-modal alignment/synchronization, opens still debated issues about its clinical value in neurological patients, possibly incompliant and highly variable from a clinical point of view. While several preclinical and clinical studies have investigated the sensitivity of PET tracers to track microglial (mainly TSPO ligands) and astrocytic (mainly MAOB ligands) activation, less studies have focused on MR specificity to this topic (e.g., through the assessment of diffusion properties and T2 relaxometry), and only few exploiting the integration of simultaneous hybrid acquisition. This review aims at summarizing and critically review the current state about PET and MR imaging for glial targets, as well as the potential added value of hybrid scanners for characterizing microglial and astrocytic activation.
Collapse
|
36
|
Prospects and challenges of imaging neuroinflammation beyond TSPO in Alzheimer's disease. Eur J Nucl Med Mol Imaging 2019; 46:2831-2847. [PMID: 31396666 PMCID: PMC6879435 DOI: 10.1007/s00259-019-04462-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 07/24/2019] [Indexed: 02/06/2023]
Abstract
Neuroinflammation, as defined by the activation of microglia and astrocytes, has emerged in the last years as a key element of the pathogenesis of neurodegenerative diseases based on genetic findings and preclinical and human studies. This has raised the need for new methodologies to assess and follow glial activation in patients, prompting the development of PET ligands for molecular imaging of glial cells and novel structural MRI and DTI tools leading to a multimodal approach. The present review describes the recent advancements in microglia and astrocyte biology in the context of health, ageing, and Alzheimer's disease, the most common dementia worldwide. The review further delves in molecular imaging discussing the challenges associated with past and present targets, including conflicting findings, and finally, presenting novel methodologies currently explored to improve our in vivo knowledge of the neuroinflammatory patterns in Alzheimer's disease. With glial cell activation as a potential therapeutic target in neurodegenerative diseases, the translational research between cell biologists, chemists, physicists, radiologists, and neurologists should be strengthened.
Collapse
|
37
|
Restoring synaptic function through multimodal therapeutics. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 168:257-275. [PMID: 31699320 DOI: 10.1016/bs.pmbts.2019.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Alzheimer's disease (AD) is the major form of dementia and a growing epidemic for which no disease-modifying treatments exist. AD is characterized by the early loss of synapses in the brain and, at later stages, neuronal death accompanied with progressive loss of cognitive functions. Here we focus on the mechanisms involved in the maintenance of the synapse and how their perturbation leads to synaptic loss. We suggest treatment strategies that particularly target energy metabolism in terms of cholesterol and glucose biochemistry in neurons and astrocytes We also discuss the potential of restoring impaired protein homeostasis through autophagy. These pathways are analyzed from a basic science perspective and suggest new avenues for discovery. We also propose several targets for both basic and translational therapeutics in these pathways and provide perspective on future AD treatment.
Collapse
|