1
|
Vanderlinden G, Vandenberghe R, Vandenbulcke M, Van Laere K. The Current Role of Tau PET Imaging in Neurodegeneration. Semin Nucl Med 2025:S0001-2998(25)00031-5. [PMID: 40263023 DOI: 10.1053/j.semnuclmed.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/24/2025]
Abstract
Neurodegenerative tauopathies are characterized by the pathological hyperphosphorylation of tau proteins that subsequently form aggregates. Tau PET tracers with affinity to bind these pathological tau aggregates have been developed to measure disease progression and to support therapeutic drug development. In this review, we summarize the pathophysiology of tau throughout the range of neurodegenerative tauopathies. We outline the available first- and second-generation tau PET tracers, with a focus on new tau PET tracer developments, and discuss the quantification of tau PET images. Next, we summarize how tau PET relates to cerebrospinal fluid and plasma tau biomarkers. Finally, we review the current recommendations on the clinical use of tau PET versus fluid tau biomarkers in diagnosis, prognosis and treatment development.
Collapse
Affiliation(s)
- Greet Vanderlinden
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Department of Neurology, University Hospitals UZ Leuven, Leuven, Belgium; Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Mathieu Vandenbulcke
- Research Group Psychiatry, Leuven Brain Institute, KU Leuven, Leuven, Belgium; Department of Geriatric Psychiatry, University Hospitals UZ Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven, Belgium; Division of Nuclear Medicine, University Hospitals UZ Leuven, Leuven, Belgium.
| |
Collapse
|
2
|
Nie P, Wu Y, Robinson J, Mekala S, Lee VMY, Li YM. In Situ Labeling of Pathogenic Tau Using Photo-Affinity Chemical Probes. ACS Chem Biol 2025; 20:581-591. [PMID: 40079621 DOI: 10.1021/acschembio.5c00073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Tau aggregation plays a crucial role in the development of Alzheimer's disease (AD). Developing specific techniques that can isolate pathogenic tau from brain tissue is important for understanding tauopathies and advancing targeted therapies. Here, we develop photoaffinity small molecular probes and a novel method for in situ tissue labeling and investigate their activity in interacting with tau in cells and AD patient brains. Based on the reported chemical structures of tau PET tracers, we designed and synthesized two tau-specific probes, namely, Tau-2 and Tau-4. After validation in cell, mouse model, and patient brain samples, our photolabeling results suggested that Tau-2 effectively labels soluble tau in cell and mouse models, while Tau-4 selectively binds high-molecular-weight tau aggregates in late-stage AD patient brain tissues. Proteomic analysis verified the specific isolation of pathogenic tau from AD brain samples. Collectively, these findings underscore the potential of our photoaffinity probes as powerful tools for investigating tau proteins and neurofibrillary tangles in neurodegenerative diseases.
Collapse
Affiliation(s)
- Pengju Nie
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, United States
| | - You Wu
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, United States
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - John Robinson
- Department of Pathology and Laboratory Medicine, Institute on Aging, and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Shekar Mekala
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, United States
| | - Virginia M Y Lee
- Department of Pathology and Laboratory Medicine, Institute on Aging, and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, United States
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| |
Collapse
|
3
|
Dybing KM, Vetter CJ, Dempsey DA, Chaudhuri S, Saykin AJ, Risacher SL. Traumatic Brain Injury and Alzheimer's Disease Biomarkers: A Systematic Review of Findings from Amyloid and Tau Positron Emission Tomography. J Neurotrauma 2025; 42:333-348. [PMID: 39639808 PMCID: PMC11971548 DOI: 10.1089/neu.2024.0055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
Traumatic brain injury (TBI) has been discussed as a risk factor for Alzheimer's disease (AD) due to its association with AD risk and earlier cognitive symptom onset. However, the mechanisms behind this relationship are unclear. Some studies have suggested TBI may increase pathological protein deposition in an AD-like pattern; others have failed to find such associations. This review covers literature that uses positron emission tomography (PET) of β-amyloid (Aβ) and/or tau to examine individuals with a history of TBI who are at increased risk for AD due to age. A comprehensive literature search was conducted on January 9, 2023, and 26 resulting citations met inclusion criteria. Common methodological concerns included small samples, limited clinical detail about participants' TBI, recall bias due to reliance on self-reported TBI, and an inability to establish causation. For both Aβ and tau, results were widespread but inconsistent. The regions that showed the most compelling evidence for increased Aβ deposition were the cingulate gyrus and cuneus/precuneus. Evidence for elevated tau was strongest in the medial temporal lobe, entorhinal cortex, precuneus, and frontal, temporal, parietal, and occipital lobes. However, conflicting findings across most regions in both Aβ- and tau-PET studies indicate the critical need for future work in expanded samples and with greater clinical detail to offer a clearer picture of the relationship between TBI and protein deposition in older individuals at risk for AD.
Collapse
Affiliation(s)
- Kaitlyn M. Dybing
- Address correspondence to: Kaitlyn M. Dybing, BS, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 West 16th Street, Suite 4100, Indianapolis, IN 46202, USA,
| | - Cecelia J. Vetter
- Ruth Lilly Medical Library, Indiana University School of Medicine, Indianapolis, USA
| | | | | | - Andrew J. Saykin
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Shannon L. Risacher
- Address correspondence to: Shannon L. Risacher, PhD, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 West 16th Street, Suite 4100, Indianapolis, IN 46202, USA,
| |
Collapse
|
4
|
Alosco ML, Mejía Pérez J, Culhane JE, Shankar R, Nowinski CJ, Bureau S, Mundada N, Smith K, Amuiri A, Asken B, Groh JR, Miner A, Pettway E, Mosaheb S, Tripodis Y, Windon C, Mercier G, Stern RA, Grinberg LT, Soleimani-Meigooni DN, Christian BT, Betthauser TJ, Stein TD, McKee AC, Mathis CA, Abrahamson EE, Ikonomovic MD, Johnson SC, Mez J, La Joie R, Schonhaut D, Rabinovici GD. 18F-MK-6240 tau PET in patients at-risk for chronic traumatic encephalopathy. Mol Neurodegener 2025; 20:23. [PMID: 39994806 PMCID: PMC11852567 DOI: 10.1186/s13024-025-00808-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Molecular biomarkers of chronic traumatic encephalopathy (CTE) are lacking. We evaluated 18F-MK-6240 tau PET as a biomarker for CTE. Two studies were done: (1) 3H-MK-6240 autoradiography and an in-vitro brain homogenate binding studies on postmortem CTE tissue, (2) an in-vivo 18F-MK-6240 tau PET study in former American football players. METHODS Autoradiography and in-vitro binding studies were done using 3H-MK-6240 on frozen temporal and frontal cortex tissue from six autopsy cases with stage III CTE compared to Alzheimer's disease. Thirty male former National Football League (NFL) players with cognitive concerns (mean age = 58.9, SD = 7.8) completed tau (18F-MK-6240) and Aβ (18F-Florbetapir) PET. Controls included 39 Aβ-PET negative, cognitively normal males (mean age = 65.7, SD = 6.3). 18F-MK-6240 SUVr images were created using 70-90 min post-injection data with inferior cerebellar gray matter as the reference. We compared SUVr between players and controls using voxelwise and region-of-interest approaches. Correlations between 18F-MK-6240 SUVr and cognitive scores were tested. RESULTS All six CTE stage III cases had Braak NFT stage III but no neuritic plaques. Two had Thal Phase 1 for Aβ; one showed a laminar pattern of 3H-MK-6240 autoradiography binding in the superior temporal cortex and less so in the dorsolateral frontal cortex, corresponding to tau-immunoreactive lesions detected using the AT8 antibody (pSer202/pThr205 tau) in adjacent tissue sections. The other CTE cases had low frequencies of cortical tau-immunoreactive deposits and no well-defined autoradiography binding. In-vitro 3H-MK-6240 binding studies to CTE brain homogenates in the case with autoradiography signal indicated high binding affinity (KD = 2.0 ± 0.9 nM, Bmax = 97 ± 24 nM, n = 3). All NFL players had negative Aβ-PET. There was variable, low-to-intermediate intensity 18F-MK-6240 uptake across participants: 16 had no cortical signal, 7 had medial temporal lobe (MTL) uptake, 2 had frontal uptake, and 4 had MTL and frontal uptake. NFL players had higher SUVr in the entorhinal cortex (d = 0.86, p = 0.001), and the parahippocampal gyrus (d = 0.39, p = 0.08). Voxelwise regressions showed increased uptake in NFL players in two bilateral anterior MTL clusters (p < 0.05 FWE). Higher parahippocampal and frontal-temporal SUVrs correlated with worse memory (r = -0.38, r = -0.40) and semantic fluency (r = -0.38, r = -0.48), respectively. CONCLUSION We present evidence of 3H-MK-6240 in-vitro binding to post-mortem CTE tissue homogenates and in vivo 18F-MK-6240 PET binding in the MTL among a subset of participants. Additional studies in larger samples and PET-to-autopsy correlations are required to further elucidate the potential of 18F-MK-6240 to detect tau pathology in CTE.
Collapse
Affiliation(s)
- Michael L Alosco
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston Medical Center, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jhony Mejía Pérez
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Julia E Culhane
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ranjani Shankar
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | | | | | - Nidhi Mundada
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Karen Smith
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Alinda Amuiri
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Breton Asken
- Department of Clinical & Health Psychology, 1Florida Alzheimer's Disease Research Center, Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Jenna R Groh
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Annalise Miner
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Erika Pettway
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sydney Mosaheb
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Yorghos Tripodis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Charles Windon
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Gustavo Mercier
- Molecular Imaging and Nuclear Medicine, Boston Medical Center, Boston, MA, USA
| | - Robert A Stern
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurosurgery, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Lea T Grinberg
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - David N Soleimani-Meigooni
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Bradley T Christian
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, USA
| | - Tobey J Betthauser
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, USA
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Thor D Stein
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
- U.S.Department of Veteran Affairs, VA Boston Healthcare System, Jamaica Plain, MA, USA
- Department of Psychiatry and Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ann C McKee
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
- U.S.Department of Veteran Affairs, VA Boston Healthcare System, Jamaica Plain, MA, USA
- Department of Psychiatry and Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eric E Abrahamson
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh HS, Pittsburgh, PA, USA
| | - Milos D Ikonomovic
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh HS, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, USA
- School of Medicine and Public Health, Wisconsin Alzheimer's Institute, University of Wisconsin-Madison, Madison, USA
| | - Jesse Mez
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Renaud La Joie
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Daniel Schonhaut
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Gil D Rabinovici
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA.
- Department of Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
- University of California, San Francisco (UCSF), Memory and Aging Center MC: 1207, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA.
| |
Collapse
|
5
|
Wongso H, Harada R, Furumoto S. Current Progress and Future Directions in Non-Alzheimer's Disease Tau PET Tracers. ACS Chem Neurosci 2025; 16:111-127. [PMID: 39762194 DOI: 10.1021/acschemneuro.4c00319] [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: 01/16/2025] Open
Abstract
Alzheimer's disease (AD) and non-AD tauopathies are dominant public health issues driven by several factors, especially in the aging population. The discovery of first-generation radiotracers, including [18F]FDDNP, [11C]PBB3, [18F]flortaucipir, and the [18F]THK series, for the in vivo detection of tauopathies has marked a significant breakthrough in the fields of neuroscience and radiopharmaceuticals, creating a robust new category of labeled compounds: tau positron emission tomography (PET) tracers. Subsequently, other tau PET tracers with improved binding properties have been developed using various chemical scaffolds to target the three-repeat/four-repeat (3R/4R) tau folds in AD. In 2020, [18F]flortaucipir was approved by the U.S. Food and Drug Administration for PET imaging of tau pathology in adult patients with cognitive deficits undergoing evaluation for AD. Despite remarkable progress in the development of AD tau PET tracers, imaging agents for rare non-AD tauopathies (4R tauopathies [predominantly expressing a 4R tau isoform], involved in progressive supranuclear palsy, corticobasal degeneration, argyrophilic grain disease, and globular glial tauopathy, and 3R tauopathies [predominantly expressing a 3R tau isoform], such as Pick's disease) remain substantially underdeveloped. In this review, we discuss recent progress in tau PET tracer development, with particular emphasis on clinically validated tracers for AD and their potential use for non-AD tauopathies. Additionally, we highlight the critical need for further development of tau PET tracers specifically designed for non-AD tauopathies, an area that remains significantly underexplored despite its importance in advancing the understanding and diagnosis of these disorders.
Collapse
Affiliation(s)
- Hendris Wongso
- Research Center for Accelerator and Radioisotope Science, Tohoku University, Sendai, Miyagi 980-0845, Japan
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten 15314, Indonesia
| | - Ryuichi Harada
- Research Center for Accelerator and Radioisotope Science, Tohoku University, Sendai, Miyagi 980-0845, Japan
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan
- Division of Brain Science, Department of Aging Research and Geriatrics Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Shozo Furumoto
- Research Center for Accelerator and Radioisotope Science, Tohoku University, Sendai, Miyagi 980-0845, Japan
- Graduate School of Pharmaceutical Science, Tohoku University, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
6
|
Mekala S, Wu Y, Li YM. Strategies of positron emission tomography (PET) tracer development for imaging of tau and α-synuclein in neurodegenerative disorders. RSC Med Chem 2024:d4md00576g. [PMID: 39678127 PMCID: PMC11638850 DOI: 10.1039/d4md00576g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder, characterized by the presence of extracellular amyloid plaques consisting of β-amyloid peptides (Aβ) and intracellular neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau (pTau) protein in the brain. Genetic and animal studies strongly indicate that Aβ, tau and neuroinflammation play important roles in the pathogenesis of AD. Several staging models showed that NFTs correlated well with the disease progression. Positron emission tomography (PET) imaging has become a widely used non-invasive technique to image NFTs for early diagnosis of AD. Despite the remarkable progress made over the past few years, tau PET imaging is still challenging due to the nature of tau pathology and the technical aspects of PET imaging. Tau pathology often coexists with other proteinopathies, such as Aβ plaques and α-synuclein aggregates. Distinguishing tau-specific signals from other overlapping pathologies is difficult, especially in the context of AD, where multiple protein aggregates are present, as well as the spectrum of different tau isoforms (3R and 4R) and conformations. Moreover, tracers should ideally have optimal pharmacokinetic properties to penetrate the blood-brain barrier (BBB) while maintaining specificity, low toxicity, low non-specific binding, rapid uptake and clearance from the brain, and formation of no radiolabeled metabolites in the brain. On the other hand, Parkinson's disease (PD) is a progressive neurodegenerative movement disorder characterized by the abnormal accumulations of α-synuclein in neurons. Heterogeneity and the unclear pathogenesis of PD hinder early and accurate diagnosis of the disease for therapeutic development in clinical use. In this review, while referring to existing reviews, we focus on the design strategies and current progress in tau (NFTs) targeting new PET tracers for AD; evolution of non-AD tau targeting PET tracers for applications including progressive supranuclear paralysis (PSP) and corticobasal degeneration (CBD); new PET tracer development for α-synuclein aggregate imaging in PD and giving an outlook for future PET tracer development.
Collapse
Affiliation(s)
- Shekar Mekala
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center 1275 York Avenue New York New York 10065 USA
| | - You Wu
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center 1275 York Avenue New York New York 10065 USA
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center New York New York 10065 USA
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center 1275 York Avenue New York New York 10065 USA
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center New York New York 10065 USA
| |
Collapse
|
7
|
Solar KG, Ventresca M, Zamyadi R, Zhang J, Jetly R, Vartanian O, Rhind SG, Dunkley BT. Repetitive subconcussion results in disrupted neural activity independent of concussion history. Brain Commun 2024; 6:fcae348. [PMID: 39440300 PMCID: PMC11495223 DOI: 10.1093/braincomms/fcae348] [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: 04/16/2024] [Revised: 07/31/2024] [Accepted: 10/06/2024] [Indexed: 10/25/2024] Open
Abstract
Concussion is a public health crisis that results in a complex cascade of neurochemical changes that can have life-changing consequences. Subconcussions are generally considered less serious, but we now realize repetitive subconcussions can lead to serious neurological deficits. Subconcussions are common in contact sports and the military where certain personnel are exposed to repetitive occupational blast overpressure. Post-mortem studies show subconcussion is a better predictor than concussion for chronic traumatic encephalopathy-a progressive and fatal neurodegenerative tauopathy, only diagnosable post-mortem-thus, an in vivo biomarker would be transformative. Magnetoencephalography captures the dynamics of neuronal electrochemical action, and functional MRI shows that functional connectivity is associated with tauopathy patterns. Therefore, both imaging modalities could provide surrogate markers of tauopathy. In this cross-sectional study, we examined the effects of repetitive subconcussion on neuronal activity and functional connectivity using magnetoencephalography and functional MRI, and on neurological symptoms and mental health in a military sample. For magnetoencephalography and outcome analyses, 81 participants were split into 'high' and 'low' blast exposure groups using the generalized blast exposure value: n = 41 high blast (26.4-65.7 years; 4 females) and n = 40 low blast (28.0-63.3 years; 8 females). For functional MRI, two high blast male participants without data were excluded: n = 39 (29.6-65.7 years). Magnetoencephalography revealed disrupted neuronal activity in participants with a greater history of repetitive subconcussions, including neural slowing (higher delta activity) in right fronto-temporal lobes and subcortical regions (hippocampus, amygdala, caudate, pallidum and thalamus), and functional dysconnectivity in the posterior default mode network (lower connectivity at low and high gamma). These abnormalities were independent of concussion or traumatic stress history, and magnetoencephalography showed functional dysconnectivity not detected in functional MRI. Besides magnetoencephalography changes, those with higher blast exposure had poorer somatic and cognitive outcomes, with no blast-related differences in mental health or associations between neurological symptoms and neuronal activity. This study suggests that repetitive subconcussions have deleterious effects on brain function and that magnetoencephalography provides an avenue for both treatment targets by identifying affected brain regions and in prevention by identifying those at risk of cumulative subconcussive neurotrauma.
Collapse
Affiliation(s)
- Kevin Grant Solar
- Neurosciences and Mental Health, Hospital for Sick Children Research Institute, Toronto, ON, Canada M5G 0A4
| | - Matthew Ventresca
- Neurosciences and Mental Health, Hospital for Sick Children Research Institute, Toronto, ON, Canada M5G 0A4
| | - Rouzbeh Zamyadi
- Neurosciences and Mental Health, Hospital for Sick Children Research Institute, Toronto, ON, Canada M5G 0A4
| | - Jing Zhang
- Defence Research and Development Canada, Toronto Research Centre, Toronto, ON, Canada M3K 2C9
| | - Rakesh Jetly
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada K1A 0K6
| | - Oshin Vartanian
- Defence Research and Development Canada, Toronto Research Centre, Toronto, ON, Canada M3K 2C9
| | - Shawn G Rhind
- Defence Research and Development Canada, Toronto Research Centre, Toronto, ON, Canada M3K 2C9
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, ON, Canada M5S 2W6
| | - Benjamin T Dunkley
- Neurosciences and Mental Health, Hospital for Sick Children Research Institute, Toronto, ON, Canada M5G 0A4
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada M5G 1X8
- Department of Diagnostic and Interventional Radiology, Hospital for Sick Children, Toronto, ON, Canada M5G 1X8
- Department of Psychology, University of Nottingham, Nottingham NG7 2RD, UK
| |
Collapse
|
8
|
Blanks W, Hanshaw M, Perez-Chadid DA, Lucke-Wold B. Emerging frontiers in Chronic Traumatic Encephalopathy: early diagnosis and implications for neurotherapeutic interventions. Expert Rev Neurother 2024; 24:953-961. [PMID: 39118236 DOI: 10.1080/14737175.2024.2385952] [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: 03/02/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024]
Abstract
INTRODUCTION Chronic Traumatic Encephalopathy (CTE) is a neurodegenerative disorder associated with repetitive head trauma. Historically, the diagnosis has been primarily clinical, which has hindered definitive early diagnosis and proactive intervention. AREAS COVERED The authors analyze the recent advancements in early diagnosis of CTE by examining biomarkers, imaging, and clinical decision tools. They discuss the identification of neuropathologies - such as tau aggregates - through novel techniques ranging from blood sampling and to brain density scanning. The reader will walk away with a better understanding of current advancements in early detection and be better equipped to deal with encephalopathies secondary to trauma in clinical practice. EXPERT OPINION Tremendous progress has been made in understanding the pathophysiology of CTE. Despite these advancements, CTE treatment is still primarily symptomatic rather than underlying disease. Future research should focus on integrating current understanding of CTE pathophysiology with treatment modalities.
Collapse
Affiliation(s)
- William Blanks
- School of Medicine, West Virginia University, Morgantown, USA
| | - Marcus Hanshaw
- School of Medicine, University of Florida, Gainesville, USA
| | | | | |
Collapse
|
9
|
Vasilevskaya A, Anastassiadis C, Thapa S, Taghdiri F, Khodadadi M, Multani N, Rusjan P, Ozzoude M, Tarazi A, Mushtaque A, Wennberg R, Houle S, Green R, Colella B, Vasdev N, Blennow K, Zetterberg H, Karikari T, Sato C, Moreno D, Rogaeva E, Mikulis D, Davis KD, Tator C, Tartaglia MC. 18F-Flortaucipir (AV1451) imaging identifies grey matter atrophy in retired athletes. J Neurol 2024; 271:6068-6079. [PMID: 39037476 PMCID: PMC11377597 DOI: 10.1007/s00415-024-12573-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/06/2024] [Accepted: 07/07/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND The long-term consequences of concussions may include pathological neurodegeneration as seen in Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE). Tau-PET showed promise as a method to detect tau pathology of CTE, but more studies are needed OBJECTIVE: This study aimed (1) to assess the association of imaging evidence of tau pathology with brain volumes in retired athletes and (2) to examine the relationship between tau-PET and neuropsychological functioning. METHODS Former contact sport athletes were recruited through the Canadian Football League Alumni Association or the Canadian Concussion Centre clinic. Athletes completed MRI, [18F]flortaucipir tau-PET, and a neuropsychological battery. Memory composite was created by averaging the Rey Auditory Verbal Learning Test and Rey Visual Design Learning Test z-scores. Grey matter (GM) volumes were age/intracranial volume corrected using normal control MRIs. Tau-PET % positivity in GM was calculated as the number of positive voxels (≥ 1.3 standardized uptake value ratio (SUVR)/total voxels). RESULTS 47 retired contact sport athletes negative for AD (age:51 ± 14; concussions/athlete:15 ± 2) and 54 normal controls (age:50 ± 13) were included. Tau-PET positive voxels had significantly lower GM volumes, compared to tau-PET negative voxels (- 0.37 ± 0.41 vs. - 0.31 ± 0.37, paired p = .006). There was a significant relationship between GM tau-PET % positivity and memory composite score (r = - .366, p = .02), controlled for age, PET scanner, and PET scan duration. There was no relationship between tau-PET measures and concussion number, or years of sport played. CONCLUSION A higher tau-PET signal was associated with reduced GM volumes and lower memory scores. Tau-PET may be useful for identifying those at risk for neurodegeneration.
Collapse
Affiliation(s)
- Anna Vasilevskaya
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, 6th Floor 6KD-407, Toronto, ON, M5T 2S8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Division of Neurology, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
- Canadian Concussion Centre, Toronto Western Hospital, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Chloe Anastassiadis
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, 6th Floor 6KD-407, Toronto, ON, M5T 2S8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Division of Neurology, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Simrika Thapa
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, 6th Floor 6KD-407, Toronto, ON, M5T 2S8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Division of Neurology, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Foad Taghdiri
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, 6th Floor 6KD-407, Toronto, ON, M5T 2S8, Canada
- Division of Neurology, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
- Canadian Concussion Centre, Toronto Western Hospital, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Mozhgan Khodadadi
- Canadian Concussion Centre, Toronto Western Hospital, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Namita Multani
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, 6th Floor 6KD-407, Toronto, ON, M5T 2S8, Canada
- Division of Neurology, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Pablo Rusjan
- Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Miracle Ozzoude
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, 6th Floor 6KD-407, Toronto, ON, M5T 2S8, Canada
| | - Apameh Tarazi
- Division of Neurology, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
- Canadian Concussion Centre, Toronto Western Hospital, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Asma Mushtaque
- Division of Neurology, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Richard Wennberg
- Division of Neurology, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
- Canadian Concussion Centre, Toronto Western Hospital, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Sylvain Houle
- Brain Health Imaging Centre, Campbell Research Institute, Centre for Addiction and Mental Health, and Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Robin Green
- Canadian Concussion Centre, Toronto Western Hospital, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- KITE Research Institute, University Health Network, Toronto, ON, Canada
| | - Brenda Colella
- Canadian Concussion Centre, Toronto Western Hospital, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- KITE Research Institute, University Health Network, Toronto, ON, Canada
| | - Neil Vasdev
- Brain Health Imaging Centre, Campbell Research Institute, Centre for Addiction and Mental Health, and Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- 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, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Thomas Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Christine Sato
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, 6th Floor 6KD-407, Toronto, ON, M5T 2S8, Canada
| | - Danielle Moreno
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, 6th Floor 6KD-407, Toronto, ON, M5T 2S8, Canada
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, 6th Floor 6KD-407, Toronto, ON, M5T 2S8, Canada
| | - David Mikulis
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Canadian Concussion Centre, Toronto Western Hospital, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Division of Neuroradiology, Joint Department of Medical Imaging, University Health Network, Toronto, ON, Canada
| | - Karen Deborah Davis
- Canadian Concussion Centre, Toronto Western Hospital, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Charles Tator
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Canadian Concussion Centre, Toronto Western Hospital, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Division of Neurosurgery, Toronto Western Hospital, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, 6th Floor 6KD-407, Toronto, ON, M5T 2S8, Canada.
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
- Division of Neurology, Toronto Western Hospital, University Health Network, Toronto, ON, Canada.
- Canadian Concussion Centre, Toronto Western Hospital, Krembil Brain Institute, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
10
|
Liu K, Tao Y, Zhao Q, Xia W, Li X, Zhang S, Yao Y, Xiang H, Han C, Tan L, Sun B, Li D, Li A, Liu C. Binding adaptability of chemical ligands to polymorphic α-synuclein amyloid fibrils. Proc Natl Acad Sci U S A 2024; 121:e2321633121. [PMID: 39172784 PMCID: PMC11363296 DOI: 10.1073/pnas.2321633121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 07/17/2024] [Indexed: 08/24/2024] Open
Abstract
α-synuclein (α-syn) assembles into structurally distinct fibril polymorphs seen in different synucleinopathies, such as Parkinson's disease and multiple system atrophy. Targeting these unique fibril structures using chemical ligands holds diagnostic significance for different disease subtypes. However, the molecular mechanisms governing small molecules interacting with different fibril polymorphs remain unclear. Here, we investigated the interactions of small molecules belonging to four distinct scaffolds, with different α-syn fibril polymorphs. Using cryo-electron microscopy, we determined the structures of these molecules when bound to the fibrils formed by E46K mutant α-syn and compared them to those bound with wild-type α-syn fibrils. Notably, we observed that these ligands exhibit remarkable binding adaptability, as they engage distinct binding sites across different fibril polymorphs. While the molecular scaffold primarily steered the binding locations and geometries on specific sites, the conjugated functional groups further refined this adaptable binding by fine-tuning the geometries and binding sites. Overall, our finding elucidates the adaptability of small molecules binding to different fibril structures, which sheds light on the diagnostic tracer and drug developments tailored to specific pathological fibril polymorphs.
Collapse
Affiliation(s)
- Kaien Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai201210, China
| | - Youqi Tao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai200030, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai200240, China
| | - Qinyue Zhao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai200030, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai200240, China
| | - Wencheng Xia
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai201210, China
| | - Xiang Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai200030, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai200240, China
| | - Shenqing Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai200030, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai200240, China
| | - Yuxuan Yao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai200030, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai200240, China
| | - Huaijiang Xiang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai201210, China
| | - Chao Han
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai201210, China
| | - Li Tan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai201210, China
| | - Bo Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai201210, China
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai200030, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai200240, China
| | - Ang Li
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai200032, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai201210, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai200032, China
| |
Collapse
|
11
|
Lindberg A, Murrell E, Tong J, Mason NS, Sohn D, Sandell J, Ström P, Stehouwer JS, Lopresti BJ, Viklund J, Svensson S, Mathis CA, Vasdev N. Ligand-based design of [ 18F]OXD-2314 for PET imaging in non-Alzheimer's disease tauopathies. Nat Commun 2024; 15:5109. [PMID: 38877019 PMCID: PMC11178805 DOI: 10.1038/s41467-024-49258-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 05/30/2024] [Indexed: 06/16/2024] Open
Abstract
Positron emission tomography (PET) imaging of tau aggregation in Alzheimer's disease (AD) is helping to map and quantify the in vivo progression of AD pathology. To date, no high-affinity tau-PET radiopharmaceutical has been optimized for imaging non-AD tauopathies. Here we show the properties of analogues of a first-in-class 4R-tau lead, [18F]OXD-2115, using ligand-based design. Over 150 analogues of OXD-2115 were synthesized and screened in post-mortem brain tissue for tau affinity against [3H]OXD-2115, and in silico models were used to predict brain uptake. [18F]OXD-2314 was identified as a selective, high-affinity non-AD tau PET radiotracer with favorable brain uptake, dosimetry, and radiometabolite profiles in rats and non-human primate and is being translated for first-in-human PET studies.
Collapse
Affiliation(s)
- Anton Lindberg
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
| | - Emily Murrell
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Junchao Tong
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
| | - N Scott Mason
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel Sohn
- Oxiant Discovery, SE-15136, Södertälje, Sweden
| | | | - Peter Ström
- Novandi Chemistry AB, SE-15136, Södertälje, Sweden
| | | | - Brian J Lopresti
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Chester A Mathis
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Neil Vasdev
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada.
- Department of Psychiatry, University of Toronto, Toronto, Canada.
| |
Collapse
|
12
|
Zheng H, Sun H, Cai Q, Tai HC. The Enigma of Tau Protein Aggregation: Mechanistic Insights and Future Challenges. Int J Mol Sci 2024; 25:4969. [PMID: 38732197 PMCID: PMC11084794 DOI: 10.3390/ijms25094969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Tau protein misfolding and aggregation are pathological hallmarks of Alzheimer's disease and over twenty neurodegenerative disorders. However, the molecular mechanisms of tau aggregation in vivo remain incompletely understood. There are two types of tau aggregates in the brain: soluble aggregates (oligomers and protofibrils) and insoluble filaments (fibrils). Compared to filamentous aggregates, soluble aggregates are more toxic and exhibit prion-like transmission, providing seeds for templated misfolding. Curiously, in its native state, tau is a highly soluble, heat-stable protein that does not form fibrils by itself, not even when hyperphosphorylated. In vitro studies have found that negatively charged molecules such as heparin, RNA, or arachidonic acid are generally required to induce tau aggregation. Two recent breakthroughs have provided new insights into tau aggregation mechanisms. First, as an intrinsically disordered protein, tau is found to undergo liquid-liquid phase separation (LLPS) both in vitro and inside cells. Second, cryo-electron microscopy has revealed diverse fibrillar tau conformations associated with different neurodegenerative disorders. Nonetheless, only the fibrillar core is structurally resolved, and the remainder of the protein appears as a "fuzzy coat". From this review, it appears that further studies are required (1) to clarify the role of LLPS in tau aggregation; (2) to unveil the structural features of soluble tau aggregates; (3) to understand the involvement of fuzzy coat regions in oligomer and fibril formation.
Collapse
Affiliation(s)
| | | | | | - Hwan-Ching Tai
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| |
Collapse
|
13
|
Su Y, Protas H, Luo J, Chen K, Alosco ML, Adler CH, Balcer LJ, Bernick C, Au R, Banks SJ, Barr WB, Coleman MJ, Dodick DW, Katz DI, Marek KL, McClean MD, McKee AC, Mez J, Daneshvar DH, Palmisano JN, Peskind ER, Turner RW, Wethe JV, Rabinovici G, Johnson K, Tripodis Y, Cummings JL, Shenton ME, Stern RA, Reiman EM. Flortaucipir tau PET findings from former professional and college American football players in the DIAGNOSE CTE research project. Alzheimers Dement 2024; 20:1827-1838. [PMID: 38134231 PMCID: PMC10984430 DOI: 10.1002/alz.13602] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 10/27/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023]
Abstract
INTRODUCTION Tau is a key pathology in chronic traumatic encephalopathy (CTE). Here, we report our findings in tau positron emission tomography (PET) measurements from the DIAGNOSE CTE Research Project. METHOD We compare flortaucipir PET measures from 104 former professional players (PRO), 58 former college football players (COL), and 56 same-age men without exposure to repetitive head impacts (RHI) or traumatic brain injury (unexposed [UE]); characterize their associations with RHI exposure; and compare players who did or did not meet diagnostic criteria for traumatic encephalopathy syndrome (TES). RESULTS Significantly elevated flortaucipir uptake was observed in former football players (PRO+COL) in prespecified regions (p < 0.05). Association between regional flortaucipir uptake and estimated cumulative head impact exposure was only observed in the superior frontal region in former players over 60 years old. Flortaucipir PET was not able to differentiate TES groups. DISCUSSION Additional studies are needed to further understand tau pathology in CTE and other individuals with a history of RHI.
Collapse
Affiliation(s)
- Yi Su
- Banner Alzheimer's Institute and Arizona Alzheimer's ConsortiumPhoenixArizonaUSA
| | - Hillary Protas
- Banner Alzheimer's Institute and Arizona Alzheimer's ConsortiumPhoenixArizonaUSA
| | - Ji Luo
- Banner Alzheimer's Institute and Arizona Alzheimer's ConsortiumPhoenixArizonaUSA
| | - Kewei Chen
- Banner Alzheimer's Institute and Arizona Alzheimer's ConsortiumPhoenixArizonaUSA
| | - Michael L. Alosco
- Department of NeurologyBoston University Alzheimer's Disease Research CenterBoston University CTE CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Charles H. Adler
- Department of NeurologyMayo Clinic College of Medicine, Mayo Clinic ArizonaScottsdaleArizonaUSA
| | - Laura J. Balcer
- Departments of NeurologyNYU Grossman School of MedicineNew YorkNew YorkUSA
- Department of Population Health and OphthalmologyNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Charles Bernick
- Cleveland Clinic Lou Ruvo Center for Brain HealthLas VegasNevadaUSA
- Department of NeurologyUniversity of WashingtonSeattleWashingtonUSA
| | - Rhoda Au
- Department of NeurologyBoston University Alzheimer's Disease Research CenterBoston University CTE CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Framingham Heart StudyFraminghamMassachusettsUSA
- Slone Epidemiology Center; Departments of Anatomy & Neurobiology, Neurology, and MedicineDepartment of EpidemiologyBoston University Chobanian & Avedisian School of Medicine; Boston University School of Public HealthBostonMassachusettsUSA
| | - Sarah J. Banks
- Departments of Neuroscience and PsychiatryUniversity of CaliforniaSan DiegoCaliforniaUSA
| | - William B. Barr
- Departments of NeurologyNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Michael J. Coleman
- Departments of Psychiatry and RadiologyPsychiatry Neuroimaging LaboratoryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - David W. Dodick
- Department of NeurologyMayo Clinic College of Medicine, Mayo Clinic ArizonaScottsdaleArizonaUSA
| | - Douglas I. Katz
- Department of NeurologyBoston University Alzheimer's Disease Research CenterBoston University CTE CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Encompass Health Braintree Rehabilitation HospitalBraintreeMassachusettsUSA
| | - Kenneth L. Marek
- Institute for Neurodegenerative Disorders, Invicro, LLCNew HavenConnecticutUSA
| | - Michael D. McClean
- Department of Environmental HealthBoston University School of Public HealthBostonMassachusettsUSA
| | - Ann C. McKee
- Department of NeurologyBoston University Alzheimer's Disease Research CenterBoston University CTE CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- VA Boston Healthcare SystemBostonMassachusettsUSA
| | - Jesse Mez
- Department of NeurologyBoston University Alzheimer's Disease Research CenterBoston University CTE CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Framingham Heart StudyFraminghamMassachusettsUSA
| | - Daniel H. Daneshvar
- Department of Physical Medicine & RehabilitationMassachusetts General Hospital, Spaulding Rehabilitation Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Joseph N. Palmisano
- Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Biostatistics and Epidemiology Data Analytics Center (BEDAC), Boston University School of Public HealthBostonMassachusettsUSA
| | - Elaine R. Peskind
- Department of Psychiatry and Behavioral SciencesVA Northwest Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System; University of Washington School of MedicineSeattleWashingtonUSA
| | - Robert W. Turner
- Department of Clinical Research & LeadershipThe George Washington University School of Medicine & Health SciencesWashingtonDistrict of ColumbiaUSA
| | - Jennifer V. Wethe
- Department of Psychiatry and PsychologyMayo Clinic School of Medicine, Mayo Clinic ArizonaScottsdaleArizonaUSA
| | - Gil Rabinovici
- Department of NeurologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Keith Johnson
- Gordon Center for Medical Imaging, Mass General Research Institute, Harvard Medical SchoolBostonMassachusettsUSA
| | - Yorghos Tripodis
- Department of NeurologyBoston University Alzheimer's Disease Research CenterBoston University CTE CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Jeffrey L. Cummings
- Department of Brain HealthChambers‐Grundy Center for Transformative NeuroscienceSchool of Integrated Health Sciences, University of Nevada Las VegasLas VegasNevadaUSA
| | - Martha E. Shenton
- Departments of Psychiatry and RadiologyPsychiatry Neuroimaging LaboratoryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Robert A. Stern
- Department of NeurologyBoston University Alzheimer's Disease Research CenterBoston University CTE CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Eric M. Reiman
- Banner Alzheimer's Institute and Arizona Alzheimer's ConsortiumPhoenixArizonaUSA
- University of Arizona, Arizona State University, Translational Genomics Research InstitutePhoenixArizonaUSA
| | | |
Collapse
|
14
|
Varlow C, Mathis CA, Vasdev N. In vitro evaluation of [ 3H]PI-2620 and structural derivatives in non-Alzheimer's tauopathies. Nucl Med Biol 2024; 130-131:108891. [PMID: 38458074 DOI: 10.1016/j.nucmedbio.2024.108891] [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: 10/27/2023] [Revised: 01/28/2024] [Accepted: 02/16/2024] [Indexed: 03/10/2024]
Abstract
Alzheimer's disease (AD) and non-AD tauopathies such as chronic traumatic encephalopathy (CTE), progressive supranuclear palsy (PSP), and corticobasal degeneration (CBD) are characterized by the abnormal aggregation of three-repeat (3R) and/or four-repeat (4R) tau isoforms. Several tau-PET tracers have been applied for human imaging of AD and non-AD tauopathies including [18F]PI-2620. Our objective is to evaluate [3H]PI-2620 and two promising structural derivatives, [3H]PI-2014 and [3H]F-4, using in vitro saturation assays and competitive binding assays against new chemical entities based on this scaffold in human AD tissues for comparison with PSP, CBD and CTE tissues. Thin section autoradiography was employed to assess specific binding and distribution of [3H]PI-2620 and [3H]F-4 in fresh-frozen human post-mortem AD, PSP, CBD and CTE tissues. Immunohistochemistry was performed for phospho-tau (AT8) and 4R-tau (RD4). Homogenate filtration binding assays were performed for saturation analysis and competitive binding studies against [3H]PI-2620. All compounds bound with high affinity in AD tissue. In PSP tissue [3H]PI-2620 demonstrated the highest affinity (5.3 nM) and in CBD tissue [3H]F-4 bound with the highest affinity (9.4 nM). Over 40 fluorinated derivatives based on PI-2620 and F-4 were screened in AD and PSP tissue. Notably, compound 2 was the most potent derivative in PSP tissue (Ki = 7.3 nM). By autoradiography, [3H]PI-2620 and [3H]F-4 demonstrated positive signals similar in intensity in AD, PSP and CTE tissues that were displaced by homologous blockade. Binding of both radiotracers aligned with immunostaining for 4R-tau. This work demonstrates that [3H]PI-2620 and [3H]F-4 show promise for imaging 4R-tau aggregates in non-AD tauopathies. PI-2620 continues to serve as a structural scaffold for PET radiotracers with higher affinity for non-AD tau over AD tau.
Collapse
Affiliation(s)
- Cassis Varlow
- Institute of Medical Science, University of Toronto, ON M5S 1A8, Canada; Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada.
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh, PA 15213, USA
| | - Neil Vasdev
- Institute of Medical Science, University of Toronto, ON M5S 1A8, Canada; Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada.
| |
Collapse
|
15
|
Vanderlinden G, Michiels L, Koole M, Lemmens R, Liessens D, Van Walleghem J, Depreitere B, Vandenbulcke M, Van Laere K. Tau Imaging in Late Traumatic Brain Injury: A [ 18F]MK-6240 Positron Emission Tomography Study. J Neurotrauma 2024; 41:420-429. [PMID: 38038357 DOI: 10.1089/neu.2023.0085] [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: 12/02/2023] Open
Abstract
Epidemiological studies have identified prior traumatic brain injury (TBI) as a risk factor for developing Alzheimer's disease (AD). Neurofibrillary tangles (NFTs) are common to AD and chronic traumatic encephalopathy following repetitive mild TBI. However, it is unclear if a single TBI is sufficient to cause accumulation of NFTs. We performed a [18F]MK-6240 positron emission tomography (PET) imaging study to assess NFTs in patients who had sustained a single TBI at least 2 years prior to study inclusion. Fourteen TBI patients (49 ± 20 years; 5 M/9 F; 8 moderate-severe, 1 mild-probable, 5 symptomatic-possible TBI) and 40 demographically similar controls (57 ± 19 years; 19 M/21 F) underwent simultaneous [18F]MK-6240 PET and magnetic resonance imaging (MRI) as well as neuropsychological assessment including the Cambridge Neuropsychological Test Automated Battery (CANTAB). A region-based voxelwise partial volume correction was applied, using parcels obtained by FreeSurfer v6.0, and standardized uptake value ratios (SUVR) were calculated relative to the cerebellar gray matter. Group differences were assessed on both a voxel- and a volume-of-interest-based level and correlations of [18F]MK-6240 SUVR with time since injury as well as with clinical outcomes were calculated. Visual assessment of TBI images did not show global or focal increases in tracer uptake in any subject. On a group level, [18F]MK-6240 SUVR was not significantly different in patients versus controls or between subgroups of moderate-severe TBI versus less severe TBI. Within the TBI group, One Touch Stockings problem solving and spatial working memory (executive function), reaction time (attention), and Mini-Mental State Examination (MMSE) (global cognition) were associated with [18F]MK-6240 SUVR. We found no group-based increase of [18F]MK-6240 brain uptake in patients scanned at least 2 years after a single TBI compared with healthy volunteers, which suggests that no NFTs are building up in the first years after a single TBI. Nonetheless, correlations with cognitive outcomes were found that warrant further investigation.
Collapse
Affiliation(s)
- Greet Vanderlinden
- Nuclear Medicine and Molecular Imaging, Imaging and Pathology, and Departments of Research Group Psychiatry, KU Leuven, Leuven, Belgium
| | - Laura Michiels
- Leuven Brain Institute, Leuven, Belgium
- Department of Neurology, University Hospitals UZ Leuven, Leuven, Belgium
- VIB, Center for Brain and Disease Research, Laboratory of Neurobiology, Belgium
- Neurosciences, and Research Group Psychiatry, KU Leuven, Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, Imaging and Pathology, and Departments of Research Group Psychiatry, KU Leuven, Leuven, Belgium
| | - Robin Lemmens
- Leuven Brain Institute, Leuven, Belgium
- Department of Neurology, University Hospitals UZ Leuven, Leuven, Belgium
- VIB, Center for Brain and Disease Research, Laboratory of Neurobiology, Belgium
- Neurosciences, and Research Group Psychiatry, KU Leuven, Leuven, Belgium
| | - Dirk Liessens
- Department of Geriatric Psychiatry, University Hospitals UZ Leuven, Leuven, Belgium
| | | | - Bart Depreitere
- Department of Neurosurgery, and University Hospitals UZ Leuven, Leuven, Belgium
| | - Mathieu Vandenbulcke
- Leuven Brain Institute, Leuven, Belgium
- Department of Geriatric Psychiatry, University Hospitals UZ Leuven, Leuven, Belgium
- Neuropsychiatry, Research Group Psychiatry, KU Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Imaging and Pathology, and Departments of Research Group Psychiatry, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, Leuven, Belgium
- Department of Nuclear Medicine, University Hospitals UZ Leuven, Leuven, Belgium
| |
Collapse
|
16
|
Aguero C, Dhaynaut M, Amaral AC, Moon SH, Neelamegam R, Scapellato M, Carazo-Casas C, Kumar S, El Fakhri G, Johnson K, Frosch MP, Normandin MD, Gómez-Isla T. Head-to-head comparison of [ 18F]-Flortaucipir, [ 18F]-MK-6240 and [ 18F]-PI-2620 postmortem binding across the spectrum of neurodegenerative diseases. Acta Neuropathol 2024; 147:25. [PMID: 38280071 PMCID: PMC10822013 DOI: 10.1007/s00401-023-02672-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 01/29/2024]
Abstract
We and others have shown that [18F]-Flortaucipir, the most validated tau PET tracer thus far, binds with strong affinity to tau aggregates in Alzheimer's (AD) but has relatively low affinity for tau aggregates in non-AD tauopathies and exhibits off-target binding to neuromelanin- and melanin-containing cells, and to hemorrhages. Several second-generation tau tracers have been subsequently developed. [18F]-MK-6240 and [18F]-PI-2620 are the two that have garnered most attention. Our recent data indicated that the binding pattern of [18F]-MK-6240 closely parallels that of [18F]-Flortaucipir. The present study aimed at the direct comparison of the autoradiographic binding properties and off-target profile of [18F]-Flortaucipir, [18F]-MK-6240 and [18F]-PI-2620 in human tissue specimens, and their potential binding to monoamine oxidases (MAO). Phosphor-screen and high resolution autoradiographic patterns of the three tracers were studied in the same postmortem tissue material from AD and non-AD tauopathies, cerebral amyloid angiopathy, synucleopathies, transactive response DNA-binding protein 43 (TDP-43)-frontotemporal lobe degeneration and controls. Our results show that the three tracers show nearly identical autoradiographic binding profiles. They all strongly bind to neurofibrillary tangles in AD but do not seem to bind to a significant extent to tau aggregates in non-AD tauopathies pointing to their limited utility for the in vivo detection of non-AD tau lesions. None of them binds to lesions containing β-amyloid, α-synuclein or TDP-43 but they all show strong off-target binding to neuromelanin and melanin-containing cells, as well as weaker binding to areas of hemorrhage. The autoradiographic binding signals of the three tracers are only weakly displaced by competing concentrations of selective MAO-B inhibitor deprenyl but not by MAO-A inhibitor clorgyline suggesting that MAO enzymes do not appear to be a significant binding target of any of them. These findings provide relevant insights for the correct interpretation of the in vivo behavior of these three tau PET tracers.
Collapse
Affiliation(s)
- Cinthya Aguero
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Maeva Dhaynaut
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Ana C Amaral
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - S-H Moon
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Ramesh Neelamegam
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Margaret Scapellato
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Carlos Carazo-Casas
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Sunny Kumar
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Keith Johnson
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Matthew P Frosch
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| | - Marc D Normandin
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Teresa Gómez-Isla
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA.
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA.
| |
Collapse
|
17
|
Dybing KM, Vetter CJ, Dempsey DA, Chaudhuri S, Saykin AJ, Risacher SL. Traumatic brain injury and Alzheimer's Disease biomarkers: A systematic review of findings from amyloid and tau positron emission tomography (PET). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.30.23298528. [PMID: 38077068 PMCID: PMC10705648 DOI: 10.1101/2023.11.30.23298528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Traumatic brain injury (TBI) has been discussed as a risk factor for Alzheimer's disease (AD) due to its association with dementia risk and earlier cognitive symptom onset. However, the mechanisms behind this relationship are unclear. Some studies have suggested TBI may increase pathological protein deposition in an AD-like pattern; others have failed to find such associations. This review covers literature that uses positron emission tomography (PET) of amyloid-β and/or tau to examine subjects with history of TBI who are at risk for AD due to advanced age. A comprehensive literature search was conducted on January 9, 2023, and 24 resulting citations met inclusion criteria. Common methodological concerns included small samples, limited clinical detail about subjects' TBI, recall bias due to reliance on self-reported TBI, and an inability to establish causation. For both amyloid and tau, results were widespread but inconsistent. The regions which showed the most compelling evidence for increased amyloid deposition were the cingulate gyrus, cuneus/precuneus, and parietal lobe. Evidence for increased tau was strongest in the medial temporal lobe, entorhinal cortex, precuneus, and frontal, temporal, parietal, and occipital lobes. However, conflicting findings across most regions of interest in both amyloid- and tau-PET studies indicate the critical need for future work in expanded samples and with greater clinical detail to offer a clearer picture of the relationship between TBI and protein deposition in older subjects at risk for AD.
Collapse
Affiliation(s)
- Kaitlyn M. Dybing
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Cecelia J. Vetter
- Ruth Lilly Medical Library, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Desarae A. Dempsey
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Soumilee Chaudhuri
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Shannon L. Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| |
Collapse
|
18
|
Harada R, Lerdsirisuk P, Shimizu Y, Yokoyama Y, Du Y, Kudo K, Ezura M, Ishikawa Y, Iwata R, Shidahara M, Ishiki A, Kikuchi A, Hatano Y, Ishihara T, Onodera O, Iwasaki Y, Yoshida M, Taki Y, Arai H, Kudo Y, Yanai K, Furumoto S, Okamura N. Preclinical Characterization of the Tau PET Tracer [ 18F]SNFT-1: Comparison of Tau PET Tracers. J Nucl Med 2023; 64:1495-1501. [PMID: 37321821 DOI: 10.2967/jnumed.123.265593] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/03/2023] [Indexed: 06/17/2023] Open
Abstract
Tau PET tracers are expected to be sufficiently sensitive to track the progression of age-related tau pathology in the medial temporal cortex. The tau PET tracer N-(4-[18F]fluoro-5-methylpyridin-2-yl)-7-aminoimidazo[1,2-a]pyridine ([18F]SNFT-1) has been successfully developed by optimizing imidazo[1,2-a]pyridine derivatives. We characterized the binding properties of [18F]SNFT-1 using a head-to-head comparison with other reported 18F-labeled tau tracers. Methods: The binding affinity of SNFT-1 to tau, amyloid, and monoamine oxidase A and B was compared with that of the second-generation tau tracers MK-6240, PM-PBB3, PI-2620, RO6958948, JNJ-64326067, and flortaucipir. In vitro binding properties of 18F-labeled tau tracers were evaluated through the autoradiography of frozen human brain tissues from patients with diverse neurodegenerative disease spectra. Pharmacokinetics, metabolism, and radiation dosimetry were assessed in normal mice after intravenous administration of [18F]SNFT-1. Results: In vitro binding assays demonstrated that [18F]SNFT-1 possesses high selectivity and high affinity for tau aggregates in Alzheimer disease (AD) brains. Autoradiographic analysis of tau deposits in medial temporal brain sections from patients with AD showed a higher signal-to-background ratio for [18F]SNFT-1 than for the other tau PET tracers and no significant binding with non-AD tau, α-synuclein, transactiviation response DNA-binding protein-43, and transmembrane protein 106B aggregates in human brain sections. Furthermore, [18F]SNFT-1 did not bind significantly to various receptors, ion channels, or transporters. [18F]SNFT-1 showed a high initial brain uptake and rapid washout from the brains of normal mice without radiolabeled metabolites. Conclusion: These preclinical data suggest that [18F]SNFT-1 is a promising and selective tau radiotracer candidate that allows the quantitative monitoring of age-related accumulation of tau aggregates in the human brain.
Collapse
Affiliation(s)
- Ryuichi Harada
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan;
- Division of Brain Science, Department of Aging Research and Geriatric Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | | | - Yuki Shimizu
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Yuka Yokoyama
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Yiqing Du
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kaede Kudo
- Division of Brain Science, Department of Aging Research and Geriatric Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Michinori Ezura
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoichi Ishikawa
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Ren Iwata
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Miho Shidahara
- Department of Quantum Science and Energy Engineering, Tohoku University, Sendai, Japan
| | - Aiko Ishiki
- Division of Brain Science, Department of Aging Research and Geriatric Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
- Division of Community Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Akio Kikuchi
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuya Hatano
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Tomohiko Ishihara
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yasushi Iwasaki
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Japan; and
| | - Mari Yoshida
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Japan; and
| | - Yasuyuki Taki
- Division of Brain Science, Department of Aging Research and Geriatric Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Hiroyuki Arai
- Division of Brain Science, Department of Aging Research and Geriatric Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Yukitsuka Kudo
- Division of Brain Science, Department of Aging Research and Geriatric Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Kazuhiko Yanai
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Shozo Furumoto
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Nobuyuki Okamura
- Division of Brain Science, Department of Aging Research and Geriatric Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| |
Collapse
|