1
|
Macedo AC, Therriault J, Tissot C, Aumont É, Servaes S, Rahmouni N, Fernandez-Arias J, Lussier FZ, Wang YT, Ng KP, Vermeiren M, Bezgin G, Socualaya KQ, Stevenson J, Hosseini SA, Chamoun M, Ferrari-Souza JP, Ferreira PCL, Bellaver B, Leffa DT, Vitali P, Zimmer ER, Ismail Z, Pascoal TA, Gauthier S, Rosa-Neto P. Modeling the progression of neuropsychiatric symptoms in Alzheimer's disease with PET-based Braak staging. Neurobiol Aging 2024; 144:127-137. [PMID: 39326302 DOI: 10.1016/j.neurobiolaging.2024.09.009] [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: 04/18/2024] [Revised: 08/15/2024] [Accepted: 09/14/2024] [Indexed: 09/28/2024]
Abstract
In Alzheimer's disease (AD), neuropsychiatric symptoms (NPS) correlate with tau deposition in the brain. Here, we investigated the association of PET-based Braak stages with NPS and assessed whether they predict annual changes in NPS. We evaluated 231 individuals in the aging and AD continuum. Participants were assigned a Braak stage at baseline and followed for 1.97 (s.d. 0.62) years. NPS were investigated using the Mild Behavioral Impairment Checklist (MBI-C) and the Neuropsychiatric Inventory Questionnaire severity (NPI-Q-S) and distress (NPI-Q-D) scales. Multiple linear regressions (MLR) assessed the association of Braak stages with baseline NPS and the annual change in NPS scores. At baseline, stages I-II, III-IV, and V-VI were associated with higher MBI-C, NPI-Q-S, and NPI-Q-D scores. Stages V-VI were associated with a significant annual increase in MBI-C scores. These findings suggest that tau accumulation may manifest clinically with an increase in NPS, which seems to be an early event in AD pathophysiology. Moreover, PET-based Braak staging appears to be a good predictor of NPS severity progression.
Collapse
Affiliation(s)
- Arthur C Macedo
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada; Lawrence Berkeley National Laboratory, 1 Cyclotron Rd, Berkeley, CA 94720, USA
| | - Étienne Aumont
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Psychology, University of Québec at Montréal, 100 Rue Sherbrooke O, Montréal, QC H2X 3P2, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Jaime Fernandez-Arias
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Firoza Z Lussier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Psychiatry, University of Pittsburgh, 3501 Forbes Avenue, Pittsburgh, PA 15213, USA
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Kok Pin Ng
- Department of Neurology, National Neuroscience Institute, 11 Jln Tan Tock Seng, Singapore 308433, Singapore
| | - Marie Vermeiren
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Kely Quispialaya Socualaya
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Seyyed Ali Hosseini
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - João Pedro Ferrari-Souza
- Department of Psychiatry, University of Pittsburgh, 3501 Forbes Avenue, Pittsburgh, PA 15213, USA
| | - Pâmela C L Ferreira
- Department of Psychiatry, University of Pittsburgh, 3501 Forbes Avenue, Pittsburgh, PA 15213, USA
| | - Bruna Bellaver
- Department of Psychiatry, University of Pittsburgh, 3501 Forbes Avenue, Pittsburgh, PA 15213, USA
| | - Douglas Teixeira Leffa
- Department of Psychiatry, University of Pittsburgh, 3501 Forbes Avenue, Pittsburgh, PA 15213, USA
| | - Paolo Vitali
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Eduardo R Zimmer
- Department of Pharmacology, Graduate Program in Biological Sciences: Pharmacology and Therapeutics; and Biochemistry, Universidade Federal do Rio Grande do Sul, 2600 Ramiro Barcelo St, Porto Alegre, RS 90.035-003, Brazil; Brain Institute of Rio Grande do Sul, PUCRS, Av. Ipiranga, 6690, Porto Alegre, RS 90610-000, Brazil
| | - Zahinoor Ismail
- Departments of Psychiatry, Clinical Neurosciences, Community Health Sciences, and Pathology, Hotchkiss Brain Institute and O'Brien Institute of Public Health, University of Calgary, Calgary, AB, Canada; National Institute for Health and Care Research Exeter Biomedical Research Centre, University of Exeter, Exeter, UK
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh, 3501 Forbes Avenue, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, 3501 Forbes Avenue, Pittsburgh, PA 15213, USA
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada.
| |
Collapse
|
2
|
Han F, Lee J, Chen X, Ziontz J, Ward T, Landau SM, Baker SL, Harrison TM, Jagust WJ. Global brain activity and its coupling with cerebrospinal fluid flow is related to tau pathology. Alzheimers Dement 2024. [PMID: 39508716 DOI: 10.1002/alz.14296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 11/15/2024]
Abstract
INTRODUCTION Factors responsible for the deposition of pathological tau in the brain are incompletely understood. This study links macroscale tau deposition in the human brain to cerebrospinal fluid (CSF) flow dynamics using resting-state functional magnetic resonance imaging (rsfMRI). METHODS Low-frequency (< 0.1 Hz) resting-state global brain activity is coupled with CSF flow and potentially reflects CSF dynamics-related clearance. We examined the correlation between rsfMRI measures of CSF inflow and global activity (gBOLD-CSF coupling) as a predictor, interacting with amyloid beta (Aβ), of tau and cortical thickness (dependent variables) across Alzheimer's Disease Neuroimaging Initiative (ADNI) participants from cognitively unimpaired through mild cognitive impairment (MCI) and Alzheimer's disease (AD). RESULTS Tau deposition in Aβ+ participants, accompanied by cortical thinning and cognitive decline, is associated with decreased gBOLD-CSF coupling. Tau mediates the relationship between coupling and thickness. DISCUSSION Findings suggest that resting-state global brain activity and CSF movements comodulate Alzheimer's tau deposition, presumably related to CSF clearance. HIGHLIGHTS A non-invasive functional magnetic resonance imaging (fMRI) assessment of a CSF clearance-related process is carried out. Global brain activity is coupled with CSF inflow in human fMRI during resting state. Global fMRI-CSF coupling is correlated with tau in Alzheimer's disease (AD). This coupling measure is also associated with cortical thickness, mediated by tau.
Collapse
Affiliation(s)
- Feng Han
- Department of Neuroscience, University of California, Berkeley, California, USA
| | - JiaQie Lee
- Department of Neuroscience, University of California, Berkeley, California, USA
| | - Xi Chen
- Department of Neuroscience, University of California, Berkeley, California, USA
- Department of Cellular and Tissue Imaging, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Jacob Ziontz
- Department of Neuroscience, University of California, Berkeley, California, USA
| | - Tyler Ward
- Department of Neuroscience, University of California, Berkeley, California, USA
| | - Susan M Landau
- Department of Neuroscience, University of California, Berkeley, California, USA
| | - Suzanne L Baker
- Department of Cellular and Tissue Imaging, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Theresa M Harrison
- Department of Neuroscience, University of California, Berkeley, California, USA
| | - William J Jagust
- Department of Neuroscience, University of California, Berkeley, California, USA
- Department of Cellular and Tissue Imaging, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| |
Collapse
|
3
|
Li T, Liu R, He Y, Zhang B, Rui X, Yang X, Wang JZ, Zeng J, Li G, Li X, Liu GP. Overexpression of TECPR1 improved cognitive function of P301S-tau mice via activation of autophagy in the early and late process. Aging Cell 2024:e14404. [PMID: 39511758 DOI: 10.1111/acel.14404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/17/2024] [Accepted: 10/25/2024] [Indexed: 11/15/2024] Open
Abstract
Autophagy disorders in AD patients and animal models were well known, however, the effect of P301S-tau on autophagy is not clear. Here, we found that autophagy related protein Tectonin Beta-Propeller Repeat-Containing Protein 1 (TECPR1) decreased in the hippocampus of P301S-tau transgenic mice by proteomics, which was proved in vivo and in vitro, and P301S-tau induced autophagic deficits in early and late process. TECPR1 overexpression attenuated P301S-tau induced autophagy defects via promoting autophagosome generation and autophagosome and lysosomes fusion. We also found that TECPR1 overexpression ameliorated the behavior disorders of P301S-tau mice with promoting tau degradation, improving synaptic plasticity and neuron loss. Lastly, CQ or 3-MA treatment reversed TECPR1 induced improvement effects on autophagic and cognitive disorders, further proved that, TECPR1 activated the early and late process of autophagy to ameliorate the cognition of P301S-tau mice. Our data suggest that TECPR1 is a potential therapy target for AD.
Collapse
Affiliation(s)
- Ting Li
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruijuan Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ye He
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingge Zhang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuexiang Rui
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Subject of Modern Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Zeng
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gang Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Li
- Department of Pathology, Wuhan No. 1 Hospital, Wuhan, China
| | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Subject of Modern Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
4
|
Ghetti B, Schweighauser M, Jacobsen MH, Gray D, Bacioglu M, Murzin AG, Glazier BS, Katsinelos T, Vidal R, Newell KL, Gao S, Garringer HJ, Spillantini MG, Scheres SHW, Goedert M. TMEM106B amyloid filaments in the Biondi bodies of ependymal cells. Acta Neuropathol 2024; 148:60. [PMID: 39503754 PMCID: PMC11541264 DOI: 10.1007/s00401-024-02807-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/11/2024] [Accepted: 09/15/2024] [Indexed: 11/09/2024]
Abstract
Biondi bodies are filamentous amyloid inclusions of unknown composition in ependymal cells of the choroid plexuses, ependymal cells lining cerebral ventricles and ependymal cells of the central canal of the spinal cord. Their formation is age-dependent and they are commonly associated with a variety of neurodegenerative conditions, including Alzheimer's disease and Lewy body disorders. Here, we show that Biondi bodies are strongly immunoreactive with TMEM239, an antibody specific for inclusions of transmembrane protein 106B (TMEM106B). Biondi bodies were labelled by both this antibody and the amyloid dye pFTAA. Many Biondi bodies were also labelled for TMEM106B and the lysosomal markers Hexosaminidase A and Cathepsin D. By transmission immuno-electron microscopy, Biondi bodies of choroid plexuses were decorated by TMEM239 and were associated with structures that resembled residual bodies or secondary lysosomes. By electron cryo-microscopy, TMEM106B filaments from Biondi bodies of choroid plexuses were similar (Biondi variant), but not identical, to the fold I that was previously identified in filaments from brain parenchyma.
Collapse
Affiliation(s)
- Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, USA.
| | | | - Max H Jacobsen
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, USA
| | - Derrick Gray
- Center for Electron Microscopy, Indiana University School of Medicine, Indianapolis, USA
| | - Mehtap Bacioglu
- Department of Clinical Neurosciences, Cambridge University, Cambridge, UK
| | - Alexey G Murzin
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Bradley S Glazier
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, USA
| | | | - Ruben Vidal
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, USA
| | - Kathy L Newell
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, USA
| | - Sujuan Gao
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, USA
| | - Holly J Garringer
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, USA
| | | | - Sjors H W Scheres
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Michel Goedert
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| |
Collapse
|
5
|
Samelson AJ, Ariqat N, McKetney J, Rohanitazangi G, Parra Bravo C, Bose R, Travaglini KJ, Lam VL, Goodness D, Dixon G, Marzette E, Jin J, Tian R, Tse E, Abskharon R, Pan H, Carroll EC, Lawrence RE, Gestwicki JE, Eisenberg D, Kanaan NM, Southworth DR, Gross JD, Gan L, Swaney DL, Kampmann M. CRISPR screens in iPSC-derived neurons reveal principles of tau proteostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.16.545386. [PMID: 37398204 PMCID: PMC10312804 DOI: 10.1101/2023.06.16.545386] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Aggregation of the protein tau defines tauopathies, which include Alzheimer's disease and frontotemporal dementia. Specific neuronal subtypes are selectively vulnerable to tau aggregation and subsequent dysfunction and death, but the underlying mechanisms are unknown. To systematically uncover the cellular factors controlling the accumulation of tau aggregates in human neurons, we conducted a genome-wide CRISPRi-based modifier screen in iPSC-derived neurons. The screen uncovered expected pathways, including autophagy, but also unexpected pathways, including UFMylation and GPI anchor synthesis. We discover that the E3 ubiquitin ligase CUL5 SOCS4 is a potent modifier of tau levels in human neurons, ubiquitinates tau, and is a correlated with vulnerability to tauopathies in mouse and human. Disruption of mitochondrial function promotes proteasomal misprocessing of tau, which generates tau proteolytic fragments like those in disease and changes tau aggregation in vitro . These results reveal new principles of tau proteostasis in human neurons and pinpoint potential therapeutic targets for tauopathies.
Collapse
|
6
|
Crawford JL, Berry AS. Examining resilience to Alzheimer's disease through the lens of monoaminergic neuromodulator systems. Trends Neurosci 2024; 47:892-903. [PMID: 39368845 PMCID: PMC11563896 DOI: 10.1016/j.tins.2024.09.004] [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: 05/20/2024] [Revised: 08/15/2024] [Accepted: 09/16/2024] [Indexed: 10/07/2024]
Abstract
The monoaminergic nuclei are thought to be some of the earliest sites of Alzheimer's disease (AD) pathology in the brain, with tau-containing pretangles appearing in these nuclei decades before the onset of clinical impairments. It has increasingly been recognized that monoamine systems represent a critical target of investigation towards understanding the progression of AD and designing early detection and treatment approaches. This review synthesizes evidence across animal studies, human neuropathology, and state-of-the-art neuroimaging and daily life assessment methods in humans, which demonstrate robust relationships between monoamine systems and AD pathophysiology and behavior. Further, the review highlights the promise of multimethod, multisystem approaches to studying monoaminergic mechanisms of resilience to AD pathology.
Collapse
Affiliation(s)
| | - Anne S Berry
- Department of Psychology, Brandeis University, Waltham, MA, USA.
| |
Collapse
|
7
|
Lavrova A, Satoh R, Pham NTT, Nguyen A, Jack CR, Petersen RC, Ross RR, Dickson DW, Lowe VJ, Whitwell JL, Josephs KA. Investigating the feasibility of 18F-flortaucipir PET imaging in the antemortem diagnosis of primary age-related tauopathy (PART): An observational imaging-pathological study. Alzheimers Dement 2024. [PMID: 39417408 DOI: 10.1002/alz.14301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/08/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024]
Abstract
INTRODUCTION Primary age-related tauopathy (PART) is characterized by neurofibrillary tangles and minimal β-amyloid deposition, diagnosed postmortem. This study investigates 18F-flortaucipir (FTP) PET imaging for antemortem PART diagnosis. METHODS We analyzed FTP PET scans from 50 autopsy-confirmed PART and 13 control subjects. Temporal lobe uptake was assessed both qualitatively and quantitatively. Demographic and clinicopathological characteristics and voxel-level uptake using SPM12 were compared between FTP-positive and FTP-negative cases. Intra-reader reproducibility was evaluated with Krippendorff's alpha. RESULTS Minimal/mild and moderate FTP uptake was seen in 32% of PART cases and 62% of controls, primarily in the left inferior temporal lobe. No demographic or clinicopathological differences were found between FTP-positive and FTP-negative cases. High intra-reader reproducibility (α = 0.83) was noted. DISCUSSION FTP PET imaging did not show a specific uptake pattern for PART diagnosis, indicating that in vivo PART identification using FTP PET is challenging. Similar uptake in controls suggests non-specific uptake in PART. HIGHLIGHTS 18F-flortaucipir (FTP) PET scans were analyzed for diagnosing PART antemortem. 32% of PART cases had minimal/mild FTP uptake in the left inferior temporal lobe. Similar to PART FTP uptake was found in 62% of control subjects. No specific uptake pattern was found, challenging in vivo PART diagnosis.
Collapse
Affiliation(s)
- Anna Lavrova
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ryota Satoh
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Aivi Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Reichard R Ross
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Keith A Josephs
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
8
|
Jagust WJ, Koeppe RA, Rabinovici GD, Villemagne VL, Harrison TM, Landau SM. The ADNI PET Core at 20. Alzheimers Dement 2024; 20:7340-7349. [PMID: 39108002 PMCID: PMC11485322 DOI: 10.1002/alz.14165] [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: 04/30/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 10/18/2024]
Abstract
The Alzheimer's Disease Neuroimaging Initiative (ADNI) PET Core has evolved over time, beginning with positron emission tomography (PET) imaging of a subsample of participants with [18F]fluorodeoxyglucose (FDG)-PET, adding tracers for measurement of β-amyloid, followed by tau tracers. This review examines the evolution of the ADNI PET Core, the novel aspects of PET imaging in each stage of ADNI, and gives an accounting of PET images available in the ADNI database. The ADNI PET Core has been and continues to be a rich resource that provides quantitative PET data and preprocessed PET images to the scientific community, allowing interrogation of both basic and clinically relevant questions. By standardizing methods across different PET scanners and multiple PET tracers, the Core has demonstrated the feasibility of large-scale, multi-center PET studies. Data managed and disseminated by the PET Core has been critical to defining pathophysiological models of Alzheimer's disease (AD) and helped to drive methods used in modern therapeutic trials. HIGHLIGHTS: The ADNI PET Core began with FDG-PET and now includes three amyloid and three tau PET ligands. The PET Core has standardized acquisition and analysis of multitracer PET images. The ADNI PET Core helped to develop methods that have facilitated clinical trials in AD.
Collapse
Affiliation(s)
- William J. Jagust
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Robert A. Koeppe
- Department of RadiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Gil D. Rabinovici
- Department of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | | | | | - Susan M. Landau
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
| | | |
Collapse
|
9
|
Parent JH, Cassady K, Jagust WJ, Berry AS. Pathological and neurochemical correlates of locus coeruleus functional network activity. Biol Psychol 2024; 192:108847. [PMID: 39038634 PMCID: PMC11464174 DOI: 10.1016/j.biopsycho.2024.108847] [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: 03/19/2024] [Revised: 06/25/2024] [Accepted: 07/15/2024] [Indexed: 07/24/2024]
Abstract
The locus coeruleus (LC) produces the neuromodulators norepinephrine and dopamine, and projects widely to subcortical and cortical brain regions. The LC has been a focus of neuroimaging biomarker development for the early detection of Alzheimer's disease (AD) since it was identified as one of the earliest brain regions to develop tau pathology. Our recent research established the use of positron emission tomography (PET) to measure LC catecholamine synthesis capacity in cognitively unimpaired older adults. We extend this work by investigating the possible influence of pathology and LC neurochemical function on LC network activity using functional magnetic resonance imaging (fMRI). In separate sessions, participants underwent PET imaging to measure LC catecholamine synthesis capacity ([18F]Fluoro-m-tyrosine), tau pathology ([18F]Flortaucipir), and amyloid-β pathology ([11C]Pittsburgh compound B), and fMRI imaging to measure LC functional network activity at rest. Consistent with a growing body of research in aging and preclinical AD, we find that higher functional network activity is associated with higher tau burden in individuals at risk of developing AD (amyloid-β positive). Critically, relationships between higher LC network activity and higher pathology (amyloid-β and tau) were moderated by LC catecholamine synthesis capacity. High levels of LC catecholamine synthesis capacity reduced relationships between higher network activity and pathology. Broadly, these findings support the view that individual differences in functional network activity are shaped by interactions between pathology and neuromodulator function, and point to catecholamine systems as potential therapeutic targets.
Collapse
Affiliation(s)
- Jourdan H Parent
- Department of Psychology, Brandeis University, Waltham, MA 02453, USA.
| | - Kaitlin Cassady
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Anne S Berry
- Department of Psychology, Brandeis University, Waltham, MA 02453, USA; Volen Center for Complex Systems, Brandeis University, Waltham, MA 02453, USA
| |
Collapse
|
10
|
Salman Y, Gérard T, Huyghe L, Colmant L, Quenon L, Malotaux V, Ivanoiu A, Lhommel R, Dricot L, Hanseeuw BJ. Amygdala atrophies in specific subnuclei in preclinical Alzheimer's disease. Alzheimers Dement 2024; 20:7205-7219. [PMID: 39254209 PMCID: PMC11485073 DOI: 10.1002/alz.14235] [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: 04/29/2024] [Revised: 07/18/2024] [Accepted: 07/27/2024] [Indexed: 09/11/2024]
Abstract
INTRODUCTION Magnetic resonance imaging (MRI) segmentation algorithms make it possible to study detailed medial temporal lobe (MTL) substructures as hippocampal subfields and amygdala subnuclei, offering opportunities to develop biomarkers for preclinical Alzheimer's disease (AD). METHODS We identified the MTL substructures significantly associated with tau-positron emission tomography (PET) signal in 581 non-demented individuals from the Alzheimer's Disease Neuroimaging Initiative (ADNI-3). We confirmed our results in our UCLouvain cohort including 110 non-demented individuals by comparing volumes between individuals with different visual Braak's stages and clinical diagnosis. RESULTS Four amygdala subnuclei (cortical, central, medial, and accessory basal) were associated with tau in amyloid beta-positive (Aβ+) clinically normal (CN) individuals, while the global amygdala and hippocampal volumes were not. Using UCLouvain data, we observed that both Braak I-II and Aβ+ CN individuals had smaller volumes in these subnuclei, while no significant difference was observed in the global structure volumes or other subfields. CONCLUSION Measuring specific amygdala subnuclei, early atrophy may serve as a marker of temporal tauopathy in preclinical AD, identifying individuals at risk of progression. HIGHLIGHTS Amygdala atrophy is not homogeneous in preclinical Alzheimer's disease (AD). Tau pathology is associated with atrophy of specific amygdala subnuclei, specifically, the central, medial, cortical, and accessory basal subnuclei. Hippocampal and amygdala volume is not associated with tau in preclinical AD. Hippocampus and CA1-3 volume is reduced in preclinical AD, regardless of tau.
Collapse
Affiliation(s)
- Yasmine Salman
- Louvain Aging Brain LabInstitute of NeuroscienceUCLouvainBrusselsBelgium
| | - Thomas Gérard
- Louvain Aging Brain LabInstitute of NeuroscienceUCLouvainBrusselsBelgium
- Nuclear Medicine DepartmentSaint‐Luc University HospitalBrusselsBelgium
| | - Lara Huyghe
- Louvain Aging Brain LabInstitute of NeuroscienceUCLouvainBrusselsBelgium
| | - Lise Colmant
- Louvain Aging Brain LabInstitute of NeuroscienceUCLouvainBrusselsBelgium
- Neurology DepartmentSaint‐Luc University HospitalBrusselsBelgium
| | - Lisa Quenon
- Louvain Aging Brain LabInstitute of NeuroscienceUCLouvainBrusselsBelgium
- Neurology DepartmentSaint‐Luc University HospitalBrusselsBelgium
| | - Vincent Malotaux
- Louvain Aging Brain LabInstitute of NeuroscienceUCLouvainBrusselsBelgium
- Department of PsychiatryMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Adrian Ivanoiu
- Neurology DepartmentSaint‐Luc University HospitalBrusselsBelgium
| | - Renaud Lhommel
- Nuclear Medicine DepartmentSaint‐Luc University HospitalBrusselsBelgium
| | - Laurence Dricot
- Louvain Aging Brain LabInstitute of NeuroscienceUCLouvainBrusselsBelgium
| | - Bernard J. Hanseeuw
- Louvain Aging Brain LabInstitute of NeuroscienceUCLouvainBrusselsBelgium
- Neurology DepartmentSaint‐Luc University HospitalBrusselsBelgium
- WELBIO DepartmentWEL Research InstituteWavreBelgium
- Department of RadiologyMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | | |
Collapse
|
11
|
Kunach P, Vaquer-Alicea J, Smith MS, Monistrol J, Hopewell R, Moquin L, Therriault J, Tissot C, Rahmouni N, Massarweh G, Soucy JP, Guiot MC, Shoichet BK, Rosa-Neto P, Diamond MI, Shahmoradian SH. Cryo-EM structure of Alzheimer's disease tau filaments with PET ligand MK-6240. Nat Commun 2024; 15:8497. [PMID: 39353896 PMCID: PMC11445244 DOI: 10.1038/s41467-024-52265-x] [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: 10/11/2023] [Accepted: 08/28/2024] [Indexed: 10/03/2024] Open
Abstract
Positron Emission Tomography (PET) ligands have advanced Alzheimer's disease (AD) diagnosis and treatment. Using autoradiography and cryo-EM, we identify AD brain tissue with elevated tau burden, purify filaments, and determine the structure of second-generation high avidity PET ligand MK-6240 at 2.31 Å resolution, which bound at a 1:1 ratio within the cleft of tau paired-helical filament (PHF), engaging with glutamine 351, lysine 353, and isoleucine 360. This information elucidates the basis of MK-6240 PET in quantifying PHF deposits in AD and may facilitate the structure-based design of superior ligands against tau amyloids.
Collapse
Affiliation(s)
- Peter Kunach
- Department of Neurology, McGill University, Montreal, QC, Canada
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, Dallas, TX, US
| | - Jaime Vaquer-Alicea
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, Dallas, TX, US
| | - Matthew S Smith
- Department of Pharmaceutical Chemistry, UCSF, San Francisco, CA, US
- Program of Biophysics, UCSF, San Francisco, CA, US
| | - Jim Monistrol
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, Dallas, TX, US
| | | | - Luc Moquin
- Montreal Neurological Institute, Montreal, QC, Canada
| | | | - Cecile Tissot
- Department of Neurology, McGill University, Montreal, QC, Canada
| | - Nesrine Rahmouni
- Department of Neurology, McGill University, Montreal, QC, Canada
| | | | | | - Marie-Christine Guiot
- Department of Neurology, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute, Montreal, QC, Canada
| | - Brian K Shoichet
- Department of Pharmaceutical Chemistry, UCSF, San Francisco, CA, US
| | - Pedro Rosa-Neto
- Department of Neurology, McGill University, Montreal, QC, Canada.
| | - Marc I Diamond
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, Dallas, TX, US.
| | - Sarah H Shahmoradian
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, Dallas, TX, US.
| |
Collapse
|
12
|
Nabizadeh F. Local molecular and connectomic contributions of tau-related neurodegeneration. GeroScience 2024:10.1007/s11357-024-01339-1. [PMID: 39343862 DOI: 10.1007/s11357-024-01339-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024] Open
Abstract
Neurodegeneration in Alzheimer's disease (AD) is known to be mostly driven by tau neurofibrillary tangles. However, both tau and neurodegeneration exhibit variability in their distribution across the brain and among individuals, and the relationship between tau and neurodegeneration might be influenced by several factors. I aimed to map local molecular and connectivity characteristics that affect the association between tau pathology and neurodegeneration. The current study was conducted on the cross-sectional tau-PET and longitudinal T1-weighted MRI scan data of 186 participants from the ADNI dataset including 71 cognitively unimpaired (CU) and 115 mild cognitive impairment (MCI) individuals. Furthermore, the normative molecular profile of a region was defined using neurotransmitter receptor densities, gene expression, T1w/T2w ratio (myelination), FDG-PET (glycolytic index, glucose metabolism, and oxygen metabolism), and synaptic density. I found that the excitatory-inhibitory (E:I) ratio, myelination, synaptic density, glycolytic index, and functional connectivity are linked with deviation in the relationship between tau and neurodegeneration. Furthermore, there was spatial similarity between tau pathology and glycolytic index, synaptic density, and functional connectivity across brain regions. The current study demonstrates that the regional susceptibility to tau-related neurodegeneration is associated with specific molecular and connectomic characteristics of the affected neural systems. I found that the molecular and connectivity architecture of the human brain is linked to the different effects of tau pathology on downstream neurodegeneration.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Alzheimer's Disease Institute, Tehran, Iran.
| |
Collapse
|
13
|
Vilkaite G, Vogel J, Mattsson-Carlgren N. Integrating amyloid and tau imaging with proteomics and genomics in Alzheimer's disease. Cell Rep Med 2024; 5:101735. [PMID: 39293391 PMCID: PMC11525023 DOI: 10.1016/j.xcrm.2024.101735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/28/2024] [Accepted: 08/20/2024] [Indexed: 09/20/2024]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease and is characterized by the aggregation of β-amyloid (Aβ) and tau in the brain. Breakthroughs in disease-modifying treatments targeting Aβ bring new hope for the management of AD. But to effectively modify and someday even prevent AD, a better understanding is needed of the biological mechanisms that underlie and link Aβ and tau in AD. Developments of high-throughput omics, including genomics, proteomics, and transcriptomics, together with molecular imaging of Aβ and tau with positron emission tomography (PET), allow us to discover and understand the biological pathways that regulate the aggregation and spread of Aβ and tau in living humans. The field of integrated omics and PET studies of Aβ and tau in AD is growing rapidly. We here provide an update of this field, both in terms of biological insights and in terms of future clinical implications of integrated omics-molecular imaging studies.
Collapse
Affiliation(s)
- Gabriele Vilkaite
- Department of Clinical Sciences Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Jacob Vogel
- Department of Clinical Sciences Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden; Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden; Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
14
|
Gebre RK, Rial AM, Raghavan S, Wiste HJ, Heeman F, Costoya-Sánchez A, Schwarz CG, Spychalla AJ, Lowe VJ, Graff-Radford J, Knopman DS, Petersen RC, Schöll M, Murray ME, Jack CR, Vemuri P. Advancing Tau PET Quantification in Alzheimer Disease with Machine Learning: Introducing THETA, a Novel Tau Summary Measure. J Nucl Med 2024; 65:1473-1480. [PMID: 39054278 PMCID: PMC11372256 DOI: 10.2967/jnumed.123.267273] [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: 12/25/2023] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Alzheimer disease (AD) exhibits spatially heterogeneous 3- or 4-repeat tau deposition across participants. Our overall goal was to develop an automated method to quantify the heterogeneous burden of tau deposition into a single number that would be clinically useful. Methods: We used tau PET scans from 3 independent cohorts: the Mayo Clinic Study of Aging and Alzheimer's Disease Research Center (Mayo, n = 1,290), the Alzheimer's Disease Neuroimaging Initiative (ADNI, n = 831), and the Open Access Series of Imaging Studies (OASIS-3, n = 430). A machine learning binary classification model was trained on Mayo data and validated on ADNI and OASIS-3 with the goal of predicting visual tau positivity (as determined by 3 raters following Food and Drug Administration criteria for 18F-flortaucipir). The machine learning model used region-specific SUV ratios scaled to cerebellar crus uptake. We estimated feature contributions based on an artificial intelligence-explainable method (Shapley additive explanations) and formulated a global tau summary measure, Tau Heterogeneity Evaluation in Alzheimer's Disease (THETA) score, using SUV ratios and Shapley additive explanations for each participant. We compared the performance of THETA with that of commonly used meta-regions of interest (ROIs) using the Mini-Mental State Examination, the Clinical Dementia Rating-Sum of Boxes, clinical diagnosis, and histopathologic staging. Results: The model achieved a balanced accuracy of 95% on the Mayo test set and at least 87% on the validation sets. It classified tau-positive and -negative participants with an AUC of 1.00, 0.96, and 0.94 on the Mayo, ADNI, and OASIS-3 cohorts, respectively. Across all cohorts, THETA showed a better correlation with the Mini-Mental State Examination and the Clinical Dementia Rating-Sum of Boxes (ρ ≥ 0.45, P < 0.05) than did meta-ROIs (ρ < 0.44, P < 0.05) and discriminated between participants who were cognitively unimpaired and those who had mild cognitive impairment with an effect size of 10.09, compared with an effect size of 3.08 for meta-ROIs. Conclusion: Our proposed approach identifies positive tau PET scans and provides a quantitative summary measure, THETA, that effectively captures heterogeneous tau deposition observed in AD. The application of THETA for quantifying tau PET in AD exhibits great potential.
Collapse
Affiliation(s)
- Robel K Gebre
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Alexis Moscoso Rial
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | | | - Heather J Wiste
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Fiona Heeman
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Alejandro Costoya-Sánchez
- Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
- Nuclear Medicine Department and Molecular Imaging Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Travesía da Choupana s/n, Santiago de Compostela, Spain
| | | | | | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Ronald C Petersen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Dementia Research Centre, Institute of Neurology, University College London, London, United Kingdom; and
| | | | | | | |
Collapse
|
15
|
Qian S, Zheng Y, Jiang T, Hou J, Cao R, Cai J, Ma E, Wang W, Song W, Xie C. A Risk Variant rs6922617 in TREM Is Discrepantly Associated With Defining Neuropathological Hallmarks in the Alzheimer's Continuum. J Gerontol A Biol Sci Med Sci 2024; 79:glae185. [PMID: 39051708 DOI: 10.1093/gerona/glae185] [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: 03/05/2024] [Indexed: 07/27/2024] Open
Abstract
The single nucleotide polymorphism (SNP)-rs6922617 in the triggering receptor expressed on myeloid cells (TREM) gene cluster is a potential risk factor for Alzheimer's disease (AD). Here, we examined whether rs6922617 is associated with AD-defining neuropathological hallmarks and memory performance. We assessed the interaction between the variant rs6922617 and levels of beta-amyloid (Aβ), tau pathology, neurodegeneration, namely amyloid-tau-neurodegeneration framework, and cognition functions in 660 healthy controls, 794 mild cognitively impaired, and 272 subjects with AD. We employed linear regression and linear mixed models to examine the association. Here we find that the SNP-rs6922617 in the TREM gene cluster is associated with a higher global amyloid-ligands positron emission tomography (Aβ-PET) burden and lower fluorodeoxyglucose positron emission tomography (FDG-PET) load. Interestingly, rs6922617 risk allele carriers exhibit a significantly reduced tau accumulation compared to the non-carriers, indicating a discrepant association with Aβ and tau pathologies. Though the participants carrying the rs6922617 risk allele do not show a correlation with poorer cognitive performance, stronger neuropathological phenotypes, and memory impairments are evident in ApoE ε4 carriers with the rs6922617 risk allele. These results support the notion that the SNP-rs6922617 in the TREM gene cluster is associated with AD-related neuropathological hallmarks, such as Aβ and FDG-mediated neurodegeneration, rather than tau accumulation. Although the direct association with memory impairment in the Alzheimer's continuum remains inconclusive, our findings suggest a potential role of rs6922617 in facilitating neuropathology hallmarks.
Collapse
Affiliation(s)
- Shuangjie Qian
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi Zheng
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tao Jiang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jialong Hou
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ruixue Cao
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jinlai Cai
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Enzi Ma
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenwen Wang
- The Center of Traditional Chinese Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weihong Song
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Oujiang Laboratory, Wenzhou, Zhejiang, China
| | - Chenglong Xie
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
16
|
Leuzy A, Raket LL, Villemagne VL, Klein G, Tonietto M, Olafson E, Baker S, Saad ZS, Bullich S, Lopresti B, Bohorquez SS, Boada M, Betthauser TJ, Charil A, Collins EC, Collins JA, Cullen N, Gunn RN, Higuchi M, Hostetler E, Hutchison RM, Iaccarino L, Insel PS, Irizarry MC, Jack CR, Jagust WJ, Johnson KA, Johnson SC, Karten Y, Marquié M, Mathotaarachchi S, Mintun MA, Ossenkoppele R, Pappas I, Petersen RC, Rabinovici GD, Rosa‐Neto P, Schwarz CG, Smith R, Stephens AW, Whittington A, Carrillo MC, Pontecorvo MJ, Haeberlein SB, Dunn B, Kolb HC, Sivakumaran S, Rowe CC, Hansson O, Doré V. Harmonizing tau positron emission tomography in Alzheimer's disease: The CenTauR scale and the joint propagation model. Alzheimers Dement 2024; 20:5833-5848. [PMID: 39041435 PMCID: PMC11497758 DOI: 10.1002/alz.13908] [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: 11/07/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Tau-positron emission tomography (PET) outcome data of patients with Alzheimer's disease (AD) cannot currently be meaningfully compared or combined when different tracers are used due to differences in tracer properties, instrumentation, and methods of analysis. METHODS Using head-to-head data from five cohorts with tau PET radiotracers designed to target tau deposition in AD, we tested a joint propagation model (JPM) to harmonize quantification (units termed "CenTauR" [CTR]). JPM is a statistical model that simultaneously models the relationships between head-to-head and anchor point data. JPM was compared to a linear regression approach analogous to the one used in the amyloid PET Centiloid scale. RESULTS A strong linear relationship was observed between CTR values across brain regions. Using the JPM approach, CTR estimates were similar to, but more accurate than, those derived using the linear regression approach. DISCUSSION Preliminary findings using the JPM support the development and adoption of a universal scale for tau-PET quantification. HIGHLIGHTS Tested a novel joint propagation model (JPM) to harmonize quantification of tau PET. Units of common scale are termed "CenTauRs". Tested a Centiloid-like linear regression approach. Using five cohorts with head-to-head tau PET, JPM outperformed linearregressionbased approach. Strong linear relationship was observed between CenTauRs values across brain regions.
Collapse
|
17
|
Soleimani-Meigooni DN, Smith R, Provost K, Lesman-Segev OH, Allen IE, Chen MK, Cho H, Edwards L, Janelidze S, La Joie R, Mundada N, Ossenkoppele R, Stomrud E, Strandberg O, Strom A, Boxer AL, Dage JL, Gorno-Tempini ML, Kramer JH, Miller BL, Rojas JC, Rosen HJ, Lyoo CH, Hansson O, Rabinovici GD. Head-to-Head Comparison of Tau and Amyloid Positron Emission Tomography Visual Reads for Differential Diagnosis of Neurodegenerative Disorders: An International, Multicenter Study. Ann Neurol 2024; 96:476-487. [PMID: 38888212 PMCID: PMC11324380 DOI: 10.1002/ana.27008] [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: 02/22/2024] [Revised: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 06/20/2024]
Abstract
OBJECTIVE We compared the accuracy of amyloid and [18F]Flortaucipir (FTP) tau positron emission tomography (PET) visual reads for distinguishing patients with mild cognitive impairment (MCI) or dementia with fluid biomarker support of Alzheimer's disease (AD). METHODS Participants with FTP-PET, amyloid-PET, and diagnosis of dementia-AD (n = 102), MCI-AD (n = 41), non-AD diseases (n = 76), and controls (n = 20) were included. AD status was determined independent of PET by cerebrospinal fluid or plasma biomarkers. The mean age was 66.9 years, and 44.8% were women. Three readers interpreted scans blindly and independently. Amyloid-PET was classified as positive/negative using tracer-specific criteria. FTP-PET was classified as positive with medial temporal lobe (MTL) binding as the minimum uptake indicating AD tau (tau-MTL+), positive with posterolateral temporal or extratemporal cortical binding in an AD-like pattern (tau-CTX+), or negative. The majority of scan interpretations were used to calculate diagnostic accuracy of visual reads in detecting MCI/dementia with fluid biomarker support for AD (MCI/dementia-AD). RESULTS Sensitivity of amyloid-PET for MCI/dementia-AD was 95.8% (95% confidence interval 91.1-98.4%), which was comparable to tau-CTX+ 92.3% (86.7-96.1%, p = 0.67) and tau-MTL+ 97.2% (93.0-99.2%, p = 0.27). Specificity of amyloid-PET for biomarker-negative healthy and disease controls was 84.4% (75.5-91.0%), which was like tau-CTX+ 88.5% (80.4-94.1%, p = 0.34), and trended toward being higher than tau-MTL+ 75.0% (65.1-83.3%, p = 0.08). Tau-CTX+ had higher specificity than tau-MTL+ (p = 0.0002), but sensitivity was lower (p = 0.02), driven by decreased sensitivity for MCI-AD (80.5% [65.1-91.2] vs. 95.1% [83.5-99.4], p = 0.03). INTERPRETATION Amyloid- and tau-PET visual reads have similar sensitivity/specificity for detecting AD in cognitively impaired patients. Visual tau-PET interpretations requiring cortical binding outside MTL increase specificity, but lower sensitivity for MCI-AD. ANN NEUROL 2024;96:476-487.
Collapse
Affiliation(s)
- David N Soleimani-Meigooni
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Ruben Smith
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Karine Provost
- Department of Nuclear Medicine, University of Montreal Hospital Center, Montréal, Canada
| | - Orit H Lesman-Segev
- Department of Diagnostic Imaging, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Isabel Elaine Allen
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Miranda K Chen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Hanna Cho
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Lauren Edwards
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
- Clinical Psychology, San Diego State University & University of California, San Diego, CA, USA
| | | | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Nidhi Mundada
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands
| | - Erik Stomrud
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Amelia Strom
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
- Health Sciences and Technology, Harvard & Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Jeffrey L Dage
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Joel H Kramer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Julio C Rojas
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Howard J Rosen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Chul H Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Oskar Hansson
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund, Sweden
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| |
Collapse
|
18
|
Vanderlip CR, Taylor L, Kim S, Harris AL, Tuteja N, Meza N, Escalante YY, McMillan L, Yassa MA, Adams JN. Amyloid-β deposition in basal frontotemporal cortex is associated with selective disruption of temporal mnemonic discrimination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609449. [PMID: 39253484 PMCID: PMC11383047 DOI: 10.1101/2024.08.23.609449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Cerebral amyloid-beta (Aβ) accumulation, a hallmark pathology of Alzheimer's disease (AD), precedes clinical impairment by two to three decades. However, it is unclear whether Aβ contributes to subtle memory deficits observed during the preclinical stage. The heterogenous emergence of Aβ deposition may selectively impact certain memory domains, which rely on distinct underlying neural circuits. In this context, we tested whether specific domains of mnemonic discrimination, a neural computation essential for episodic memory, exhibit specific deficits related to early Aβ deposition. We tested 108 cognitively unimpaired human older adults (66% female) who underwent 18F-florbetapir positron emission tomography (Aβ-PET), and a control group of 35 young adults, on a suite of mnemonic discrimination tasks taxing object, spatial, and temporal domains. We hypothesized that Aβ pathology would be selectively associated with temporal discrimination performance due to Aβ's propensity to accumulate in the basal frontotemporal cortex, which supports temporal processing. Consistent with this hypothesis, we found a dissociation in which generalized age-related deficits were found for object and spatial mnemonic discrimination, while Aβ-PET levels were selectively associated with deficits in temporal mnemonic discrimination. Further, we found that higher Aβ-PET levels in medial orbitofrontal and inferior temporal cortex, regions supporting temporal processing, were associated with greater temporal mnemonic discrimination deficits, pointing to the selective vulnerability of circuits related to temporal processing early in AD progression. These results suggest that Aβ accumulation within basal frontotemporal regions may disrupt temporal mnemonic discrimination in preclinical AD, and may serve as a sensitive behavioral biomarker of emerging AD progression.
Collapse
Affiliation(s)
- Casey R. Vanderlip
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| | - Lisa Taylor
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| | - Soyun Kim
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| | - Alyssa L. Harris
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| | - Nandita Tuteja
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| | - Novelle Meza
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| | - Yuritza Y. Escalante
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| | - Liv McMillan
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| | - Michael A. Yassa
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| | - Jenna N. Adams
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| |
Collapse
|
19
|
Shoemaker RL, Larsen RJ, Larsen PA. Single-domain antibodies and aptamers drive new opportunities for neurodegenerative disease research. Front Immunol 2024; 15:1426656. [PMID: 39238639 PMCID: PMC11374656 DOI: 10.3389/fimmu.2024.1426656] [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: 05/01/2024] [Accepted: 07/22/2024] [Indexed: 09/07/2024] Open
Abstract
Neurodegenerative diseases (NDs) in mammals, such as Alzheimer's disease (AD), Parkinson's disease (PD), and transmissible spongiform encephalopathies (TSEs), are characterized by the accumulation of misfolded proteins in the central nervous system (CNS). Despite the presence of these pathogenic proteins, the immune response in affected individuals remains notably muted. Traditional immunological strategies, particularly those reliant on monoclonal antibodies (mAbs), face challenges related to tissue penetration, blood-brain barrier (BBB) crossing, and maintaining protein stability. This has led to a burgeoning interest in alternative immunotherapeutic avenues. Notably, single-domain antibodies (or nanobodies) and aptamers have emerged as promising candidates, as their reduced size facilitates high affinity antigen binding and they exhibit superior biophysical stability compared to mAbs. Aptamers, synthetic molecules generated from DNA or RNA ligands, present both rapid production times and cost-effective solutions. Both nanobodies and aptamers exhibit inherent qualities suitable for ND research and therapeutic development. Cross-seeding events must be considered in both traditional and small-molecule-based immunodiagnostic and therapeutic approaches, as well as subsequent neurotoxic impacts and complications beyond protein aggregates. This review delineates the challenges traditional immunological methods pose in ND research and underscores the potential of nanobodies and aptamers in advancing next-generation ND diagnostics and therapeutics.
Collapse
Affiliation(s)
- Rachel L Shoemaker
- Minnesota Center for Prion Research and Outreach (MNPRO), University of Minnesota, St. Paul, MN, United States
- Department of Biomedical and Veterinary Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN, United States
| | - Roxanne J Larsen
- Department of Biomedical and Veterinary Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN, United States
- Priogen Corp., St. Paul, MN, United States
| | - Peter A Larsen
- Minnesota Center for Prion Research and Outreach (MNPRO), University of Minnesota, St. Paul, MN, United States
- Department of Biomedical and Veterinary Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN, United States
| |
Collapse
|
20
|
Schworer EK, Zammit MD, Wang J, Handen BL, Betthauser T, Laymon CM, Tudorascu DL, Cohen AD, Zaman SH, Ances BM, Mapstone M, Head E, Klunk WE, Christian BT, Hartley SL. Amyloid age and tau PET timeline to symptomatic Alzheimer's disease in Down syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.08.24311702. [PMID: 39211859 PMCID: PMC11361254 DOI: 10.1101/2024.08.08.24311702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background Adults with Down syndrome (DS) are at risk for Alzheimer's disease (AD). Recent natural history cohort studies have characterized AD biomarkers, with a focus on PET amyloid-beta (Aβ) and PET tau. Leveraging these well-characterized biomarkers, the present study examined the timeline to symptomatic AD based on estimated years since reaching Aβ+, referred to as "amyloid age", and in relation to tau in a large cohort of individuals with DS. Methods In this multicenter cohort study, 25 - 57-year-old adults with DS (n = 167) were assessed twice from 2017 to 2022, with approximately 32 months between visits as part of the Alzheimer Biomarker Consortium - Down Syndrome. Adults with DS completed amyloid and tau PET scans, and were administered the modified Cued Recall Test and the Down Syndrome Mental Status Examination. Study partners completed the National Task Group-Early Detection Screen for Dementia. Findings Mixed linear regressions showed significant quadratic associations between amyloid age and cognitive performance and cubic associations between amyloid age and tau, both at baseline and across 32 months. Using broken stick regression models, differences in mCRT scores were detected beginning 2.7 years following Aβ+ in cross-sectional models, with an estimated decline of 1.3 points per year. Increases in tau began, on average, 2.7 - 6.1 years following Aβ+. On average, participants with mild cognitive impairment were 7.4 years post Aβ+ and those with dementia were 12.7 years post Aβ+. Interpretation There is a short timeline to initial cognitive decline and dementia from Aβ+ (Centiloid = 18) and tau deposition in DS relative to late onset AD. The established timeline based on amyloid age (or equivalent Centiloid values) is important for clinical practice and informing AD clinical trials, and avoids limitations of timelines based on chronological age. Funding. National Institute on Aging and the National Institute for Child Health and Human Development. Research in Context Evidence before this study: We searched PubMed for articles published involving the progression of Aβ and tau deposition in adults with Down syndrome from database inception to March 1, 2024. Terms included "amyloid", "Down syndrome", "tau", "Alzheimer's disease", "cognitive decline", and "amyloid chronicity," with no language restrictions. One previous study outlined the progression of tau in adults with Down syndrome without consideration of cognitive decline or clinical status. Other studies reported cognitive decline associated with Aβ burden and estimated years to AD symptom onset in Down syndrome. Amyloid age estimates have also been created for older neurotypical adults and compared to cognitive performance, but this has not been investigated in Down syndrome.Added value of this study: The timeline to symptomatic Alzheimer's disease in relation to amyloid, expressed as duration of Aβ+, and tau has yet to be described in adults with Down syndrome. Our longitudinal study is the first to provide a timeline of cognitive decline and transition to mild cognitive impairment and dementia in relation to Aβ+.Implications of all the available evidence: In a cohort study of 167 adults with Down syndrome, cognitive decline began 2.7 - 5.4 years and tau deposition began 2.7 - 6.1 years following Aβ+ (Centiloid = 18). Adults with Down syndrome converted to MCI after ~7 years and dementia after ~12-13 years of Aβ+. This shortened timeline to AD symptomology from Aβ+ and tau deposition in DS based on amyloid age (or corresponding Centiloid values) can inform clinical AD intervention trials and is of use in clinical settings.
Collapse
|
21
|
Sun Z, Kwon JS, Ren Y, Chen S, Walker CK, Lu X, Cates K, Karahan H, Sviben S, Fitzpatrick JAJ, Valdez C, Houlden H, Karch CM, Bateman RJ, Sato C, Mennerick SJ, Diamond MI, Kim J, Tanzi RE, Holtzman DM, Yoo AS. Modeling late-onset Alzheimer's disease neuropathology via direct neuronal reprogramming. Science 2024; 385:adl2992. [PMID: 39088624 DOI: 10.1126/science.adl2992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 05/31/2024] [Indexed: 08/03/2024]
Abstract
Late-onset Alzheimer's disease (LOAD) is the most common form of Alzheimer's disease (AD). However, modeling sporadic LOAD that endogenously captures hallmark neuronal pathologies such as amyloid-β (Aβ) deposition, tau tangles, and neuronal loss remains an unmet need. We demonstrate that neurons generated by microRNA (miRNA)-based direct reprogramming of fibroblasts from individuals affected by autosomal dominant AD (ADAD) and LOAD in a three-dimensional environment effectively recapitulate key neuropathological features of AD. Reprogrammed LOAD neurons exhibit Aβ-dependent neurodegeneration, and treatment with β- or γ-secretase inhibitors before (but not subsequent to) Aβ deposit formation mitigated neuronal death. Moreover inhibiting age-associated retrotransposable elements in LOAD neurons reduced both Aβ deposition and neurodegeneration. Our study underscores the efficacy of modeling late-onset neuropathology of LOAD through high-efficiency miRNA-based neuronal reprogramming.
Collapse
Affiliation(s)
- Zhao Sun
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ji-Sun Kwon
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Program in Computational and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yudong Ren
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Program in Developmental, Regenerative, and Stem Cell Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shawei Chen
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Courtney K Walker
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xinguo Lu
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kitra Cates
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Program in Molecular Genetics and Genomics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hande Karahan
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sanja Sviben
- Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James A J Fitzpatrick
- Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Clarissa Valdez
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Henry Houlden
- UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Celeste M Karch
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Randall J Bateman
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chihiro Sato
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven J Mennerick
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marc I Diamond
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jungsu Kim
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - David M Holtzman
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrew S Yoo
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
22
|
Fernandes S, Revanna J, Pratt J, Hayes N, Marchetto MC, Gage FH. Modeling Alzheimer's disease using human cell derived brain organoids and 3D models. Front Neurosci 2024; 18:1434945. [PMID: 39156632 PMCID: PMC11328153 DOI: 10.3389/fnins.2024.1434945] [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: 05/19/2024] [Accepted: 07/10/2024] [Indexed: 08/20/2024] Open
Abstract
Age-related neurodegenerative diseases, like Alzheimer's disease (AD), are challenging diseases for those affected with no cure and limited treatment options. Functional, human derived brain tissues that represent the diverse genetic background and cellular subtypes contributing to sporadic AD (sAD) are limited. Human stem cell derived brain organoids recapitulate some features of human brain cytoarchitecture and AD-like pathology, providing a tool for illuminating the relationship between AD pathology and neural cell dysregulation leading to cognitive decline. In this review, we explore current strategies for implementing brain organoids in the study of AD as well as the challenges associated with investigating age-related brain diseases using organoid models.
Collapse
Affiliation(s)
- Sarah Fernandes
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Jasmin Revanna
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Joshua Pratt
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Department of Biology, San Diego State University, San Diego, CA, United States
| | - Nicholas Hayes
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Department of Biological Sciences, California State University, San Marcos, CA, United States
| | - Maria C. Marchetto
- Department of Anthropology, Center for Academic Research and Training in Anthropogeny (CARTA), University of California, San Diego, La Jolla, CA, United States
| | - Fred H. Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
| |
Collapse
|
23
|
Duggan MR, Gomez GT, Joynes CM, Bilgel M, Chen J, Fattorelli N, Hohman TJ, Mancuso R, Cordon J, Castellano T, Koran MEI, Candia J, Lewis A, Moghekar A, Ashton NJ, Kac PR, Karikari TK, Blennow K, Zetterberg H, Martinez-Muriana A, De Strooper B, Thambisetty M, Ferrucci L, Gottesman RF, Coresh J, Resnick SM, Walker KA. Proteome-wide analysis identifies plasma immune regulators of amyloid-beta progression. Brain Behav Immun 2024; 120:604-619. [PMID: 38977137 DOI: 10.1016/j.bbi.2024.07.002] [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: 04/04/2024] [Revised: 06/07/2024] [Accepted: 07/04/2024] [Indexed: 07/10/2024] Open
Abstract
While immune function is known to play a mechanistic role in Alzheimer's disease (AD), whether immune proteins in peripheral circulation influence the rate of amyloid-β (Aβ) progression - a central feature of AD - remains unknown. In the Baltimore Longitudinal Study of Aging, we quantified 942 immunological proteins in plasma and identified 32 (including CAT [catalase], CD36 [CD36 antigen], and KRT19 [keratin 19]) associated with rates of cortical Aβ accumulation measured with positron emission tomography (PET). Longitudinal changes in a subset of candidate proteins also predicted Aβ progression, and the mid- to late-life (20-year) trajectory of one protein, CAT, was associated with late-life Aβ-positive status in the Atherosclerosis Risk in Communities (ARIC) study. Genetic variation that influenced plasma levels of CAT, CD36 and KRT19 predicted rates of Aβ accumulation, including causal relationships with Aβ PET levels identified with two-sample Mendelian randomization. In addition to associations with tau PET and plasma AD biomarker changes, as well as expression patterns in human microglia subtypes and neurovascular cells in AD brain tissue, we showed that 31 % of candidate proteins were related to mid-life (20-year) or late-life (8-year) dementia risk in ARIC. Our findings reveal plasma proteins associated with longitudinal Aβ accumulation, and identify specific peripheral immune mediators that may contribute to the progression of AD pathophysiology.
Collapse
Affiliation(s)
- Michael R Duggan
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Gabriela T Gomez
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cassandra M Joynes
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Jingsha Chen
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nicola Fattorelli
- VIB Center for Brain and Disease Research, Flanders Institute for Biotechnology, Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Renzo Mancuso
- Microglia and Inflammation in Neurological Disorders Laboratory, Center for Molecular Neurology, Flanders Institute for Biotechnology, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jenifer Cordon
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Tonnar Castellano
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mary Ellen I Koran
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Julián Candia
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Alexandria Lewis
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK; NIHR Biomedical Research Center for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK; Center for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Przemysław R Kac
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; ICM Institute, Pitié-Salpêtrière University Hospital, Sorbonne University, Paris, France; First Affiliated Hospital, University of Science and Technology of China, Anhui, PR China
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, University College London Institute of Neurology, London, UK; UK Dementia Research Institute, University College London, London, UK; Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong Special Administrative Region; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Anna Martinez-Muriana
- VIB Center for Brain and Disease Research, Flanders Institute for Biotechnology, Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Bart De Strooper
- VIB Center for Brain and Disease Research, Flanders Institute for Biotechnology, Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium; UK Dementia Research Institute, University College London, London, UK
| | - Madhav Thambisetty
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Rebecca F Gottesman
- Stroke Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Josef Coresh
- Departments of Population Health and Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
24
|
Jack CR, Andrews JS, Beach TG, Buracchio T, Dunn B, Graf A, Hansson O, Ho C, Jagust W, McDade E, Molinuevo JL, Okonkwo OC, Pani L, Rafii MS, Scheltens P, Siemers E, Snyder HM, Sperling R, Teunissen CE, Carrillo MC. Revised criteria for diagnosis and staging of Alzheimer's disease: Alzheimer's Association Workgroup. Alzheimers Dement 2024; 20:5143-5169. [PMID: 38934362 PMCID: PMC11350039 DOI: 10.1002/alz.13859] [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: 02/07/2024] [Revised: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 06/28/2024]
Abstract
The National Institute on Aging and the Alzheimer's Association convened three separate work groups in 2011 and single work groups in 2012 and 2018 to create recommendations for the diagnosis and characterization of Alzheimer's disease (AD). The present document updates the 2018 research framework in response to several recent developments. Defining diseases biologically, rather than based on syndromic presentation, has long been standard in many areas of medicine (e.g., oncology), and is becoming a unifying concept common to all neurodegenerative diseases, not just AD. The present document is consistent with this principle. Our intent is to present objective criteria for diagnosis and staging AD, incorporating recent advances in biomarkers, to serve as a bridge between research and clinical care. These criteria are not intended to provide step-by-step clinical practice guidelines for clinical workflow or specific treatment protocols, but rather serve as general principles to inform diagnosis and staging of AD that reflect current science. HIGHLIGHTS: We define Alzheimer's disease (AD) to be a biological process that begins with the appearance of AD neuropathologic change (ADNPC) while people are asymptomatic. Progression of the neuropathologic burden leads to the later appearance and progression of clinical symptoms. Early-changing Core 1 biomarkers (amyloid positron emission tomography [PET], approved cerebrospinal fluid biomarkers, and accurate plasma biomarkers [especially phosphorylated tau 217]) map onto either the amyloid beta or AD tauopathy pathway; however, these reflect the presence of ADNPC more generally (i.e., both neuritic plaques and tangles). An abnormal Core 1 biomarker result is sufficient to establish a diagnosis of AD and to inform clinical decision making throughout the disease continuum. Later-changing Core 2 biomarkers (biofluid and tau PET) can provide prognostic information, and when abnormal, will increase confidence that AD is contributing to symptoms. An integrated biological and clinical staging scheme is described that accommodates the fact that common copathologies, cognitive reserve, and resistance may modify relationships between clinical and biological AD stages.
Collapse
Affiliation(s)
| | - J. Scott Andrews
- Global Evidence & OutcomesTakeda Pharmaceuticals Company LimitedCambridgeMassachusettsUSA
| | - Thomas G. Beach
- Civin Laboratory for NeuropathologyBanner Sun Health Research InstituteSun CityArizonaUSA
| | - Teresa Buracchio
- Office of NeuroscienceU.S. Food and Drug AdministrationSilver SpringMarylandUSA
| | - Billy Dunn
- The Michael J. Fox Foundation for Parkinson's ResearchNew YorkNew YorkUSA
| | - Ana Graf
- NovartisNeuroscience Global Drug DevelopmentBaselSwitzerland
| | - Oskar Hansson
- Department of Clinical Sciences Malmö, Faculty of MedicineLund UniversityLundSweden
- Memory ClinicSkåne University Hospital, MalmöLundSweden
| | - Carole Ho
- DevelopmentDenali TherapeuticsSouth San FranciscoCaliforniaUSA
| | - William Jagust
- School of Public Health and Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Eric McDade
- Department of NeurologyWashington University St. Louis School of MedicineSt. LouisMissouriUSA
| | - Jose Luis Molinuevo
- Department of Global Clinical Development H. Lundbeck A/SExperimental MedicineCopenhagenDenmark
| | - Ozioma C. Okonkwo
- Department of Medicine, Division of Geriatrics and GerontologyUniversity of Wisconsin School of MedicineMadisonWisconsinUSA
| | - Luca Pani
- University of MiamiMiller School of MedicineMiamiFloridaUSA
| | - Michael S. Rafii
- Alzheimer's Therapeutic Research Institute (ATRI)Keck School of Medicine at the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Philip Scheltens
- Amsterdam University Medical Center (Emeritus)NeurologyAmsterdamthe Netherlands
| | - Eric Siemers
- Clinical ResearchAcumen PharmaceuticalsZionsvilleIndianaUSA
| | - Heather M. Snyder
- Medical & Scientific Relations DivisionAlzheimer's AssociationChicagoIllinoisUSA
| | - Reisa Sperling
- Department of Neurology, Brigham and Women's HospitalMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Charlotte E. Teunissen
- Department of Laboratory MedicineAmsterdam UMC, Neurochemistry LaboratoryAmsterdamthe Netherlands
| | - Maria C. Carrillo
- Medical & Scientific Relations DivisionAlzheimer's AssociationChicagoIllinoisUSA
| |
Collapse
|
25
|
Josephs KA, Tosakulwong N, Weigand SD, Graff-Radford J, Schwarz CG, Senjem ML, Machulda MM, Kantarci K, Knopman DS, Nguyen A, Reichard RR, Dickson DW, Petersen RC, Lowe VJ, Jack CR, Whitwell JL. Flortaucipir PET uncovers relationships between tau and amyloid-β in primary age-related tauopathy and Alzheimer's disease. Sci Transl Med 2024; 16:eado8076. [PMID: 39047115 PMCID: PMC11423951 DOI: 10.1126/scitranslmed.ado8076] [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: 02/21/2024] [Accepted: 06/28/2024] [Indexed: 07/27/2024]
Abstract
[18F]-Flortaucipir positron emission tomography (PET) is considered a good biomarker of Alzheimer's disease. However, it is unknown how flortaucipir is associated with the distribution of tau across brain regions and how these associations are influenced by amyloid-β. It is also unclear whether flortaucipir can detect tau in definite primary age-related tauopathy (PART). We identified 248 individuals at Mayo Clinic who had undergone [18F]-flortaucipir PET during life, had died, and had undergone an autopsy, 239 cases of which also had amyloid-β PET. We assessed nonlinear relationships between flortaucipir uptake in nine medial temporal and cortical regions, Braak tau stage, and Thal amyloid-β phase using generalized additive models. We found that flortaucipir uptake was greater with increasing tau stage in all regions. Increased uptake at low tau stages in medial temporal regions was only observed in cases with a high amyloid-β phase. Flortaucipir uptake linearly increased with the amyloid-β phase in medial temporal and cortical regions. The highest flortaucipir uptake occurred with high Alzheimer's disease neuropathologic change (ADNC) scores, followed by low-intermediate ADNC scores, then PART, with the entorhinal cortex providing the best differentiation between groups. Flortaucipir PET had limited ability to detect PART, and imaging-defined PART did not correspond with pathologically defined PART. In summary, spatial patterns of flortaucipir mirrored the histopathological tau distribution, were influenced by the amyloid-β phase, and were useful for distinguishing different ADNC scores and PART.
Collapse
Affiliation(s)
- Keith A Josephs
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Nirubol Tosakulwong
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Stephen D Weigand
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Matthew L Senjem
- Department of Information Technology, Mayo Clinic, Rochester, MN 55905, USA
| | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Aivi Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - R Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
26
|
Dominguez EN, Corrada MM, Kawas CH, Stark CEL. Resilience to AD pathology in Top Cognitive Performers. Front Aging Neurosci 2024; 16:1428695. [PMID: 39055052 PMCID: PMC11270559 DOI: 10.3389/fnagi.2024.1428695] [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: 05/06/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Successful cognitive aging is often thought to result from resistance to the accumulation of pathology, resilience to the effects of pathological accumulation, or some combination of the two. While evidence for resilience has been found in typical aging populations, the oldest-old provide us with a unique window into the role of pathological accumulation in impacting cognition. Here, we aimed to assess group differences in measures of amyloid and tau across older age groups using data from the Alzheimer's Disease Neuroimaging Initiative (ADNI age: 60-89) and The 90+ Study (age: 90-101). Additionally, using the ADNI dataset, we performed exploratory analyses of regional cingulate AV-45 SUVRs to assess if amyloid load in particular areas was associated with Top Cognitive Performance (TCP). Consistent with the literature, results showed no group differences in amyloid SUVRs both regionally and in the whole cortex. For tau with AV-1451, we also observed no differences in Braak composite SUVRs. Interestingly, these relationships persisted in the oldest-old. This indicates that Top Cognitive Performance throughout aging does not reflect resistance to amyloid and tau burden, but that other mechanisms may be associated with protection against amyloid and tau related neurodegeneration.
Collapse
Affiliation(s)
- Elena Nicole Dominguez
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
| | - María M. Corrada
- Department of Neurology, University of California, Irvine, Irvine, CA, United States
- Department of Epidemiology, University of California, Irvine, Irvine, CA, United States
| | - Claudia H. Kawas
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
- Department of Neurology, University of California, Irvine, Irvine, CA, United States
| | - Craig E. L. Stark
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
27
|
Murai SA, Mano T, Sanes JN, Watanabe T. Atypical intrinsic neural timescale in the left angular gyrus in Alzheimer's disease. Brain Commun 2024; 6:fcae199. [PMID: 38993284 PMCID: PMC11227993 DOI: 10.1093/braincomms/fcae199] [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: 10/15/2023] [Revised: 04/18/2024] [Accepted: 06/07/2024] [Indexed: 07/13/2024] Open
Abstract
Alzheimer's disease is characterized by cognitive impairment and progressive brain atrophy. Recent human neuroimaging studies reported atypical anatomical and functional changes in some regions in the default mode network in patients with Alzheimer's disease, but which brain area of the default mode network is the key region whose atrophy disturbs the entire network activity and consequently contributes to the symptoms of the disease remains unidentified. Here, in this case-control study, we aimed to identify crucial neural regions that mediated the phenotype of Alzheimer's disease, and as such, we examined the intrinsic neural timescales-a functional metric to evaluate the capacity to integrate diverse neural information-and grey matter volume of the regions in the default mode network using resting-state functional MRI images and structural MRI data obtained from individuals with Alzheimer's disease and cognitively typical people. After confirming the atypically short neural timescale of the entire default mode network in Alzheimer's disease and its link with the symptoms of the disease, we found that the shortened neural timescale of the default mode network was associated with the aberrantly short neural timescale of the left angular gyrus. Moreover, we revealed that the shortened neural timescale of the angular gyrus was correlated with the atypically reduced grey matter volume of this parietal region. Furthermore, we identified an association between the neural structure, brain function and symptoms and proposed a model in which the reduced grey matter volume of the left angular gyrus shortened the intrinsic neural time of the region, which then destabilized the entire neural timescale of the default mode network and resultantly contributed to cognitive decline in Alzheimer's disease. These findings highlight the key role of the left angular gyrus in the anatomical and functional aetiology of Alzheimer's disease.
Collapse
Affiliation(s)
- Shota A Murai
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study, Bunkyo City, Tokyo 113-0033, Japan
| | - Tatsuo Mano
- Department of Degenerative Neurological Diseases, National Center of Neurology and Psychiatry, Tokyo 187-8551, Japan
| | - Jerome N Sanes
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
- Center for Neurorestoration and Neurotechnology, Veterans Affairs Providence Healthcare System, Providence, RI 02908, USA
| | - Takamitsu Watanabe
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study, Bunkyo City, Tokyo 113-0033, Japan
| |
Collapse
|
28
|
Rodriguez-Rodriguez P, Arroyo-Garcia LE, Tsagkogianni C, Li L, Wang W, Végvári Á, Salas-Allende I, Plautz Z, Cedazo-Minguez A, Sinha SC, Troyanskaya O, Flajolet M, Yao V, Roussarie JP. A cell autonomous regulator of neuronal excitability modulates tau in Alzheimer's disease vulnerable neurons. Brain 2024; 147:2384-2399. [PMID: 38462574 PMCID: PMC11224620 DOI: 10.1093/brain/awae051] [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: 10/17/2023] [Revised: 01/12/2024] [Accepted: 01/19/2024] [Indexed: 03/12/2024] Open
Abstract
Neurons from layer II of the entorhinal cortex (ECII) are the first to accumulate tau protein aggregates and degenerate during prodromal Alzheimer's disease. Gaining insight into the molecular mechanisms underlying this vulnerability will help reveal genes and pathways at play during incipient stages of the disease. Here, we use a data-driven functional genomics approach to model ECII neurons in silico and identify the proto-oncogene DEK as a regulator of tau pathology. We show that epigenetic changes caused by Dek silencing alter activity-induced transcription, with major effects on neuronal excitability. This is accompanied by the gradual accumulation of tau in the somatodendritic compartment of mouse ECII neurons in vivo, reactivity of surrounding microglia, and microglia-mediated neuron loss. These features are all characteristic of early Alzheimer's disease. The existence of a cell-autonomous mechanism linking Alzheimer's disease pathogenic mechanisms in the precise neuron type where the disease starts provides unique evidence that synaptic homeostasis dysregulation is of central importance in the onset of tau pathology in Alzheimer's disease.
Collapse
Affiliation(s)
| | | | - Christina Tsagkogianni
- Department of Neurobiology Care Sciences and Society, Karolinska Institutet, 17 164, Solna, Sweden
| | - Lechuan Li
- Department of Computer Science, Rice University, Houston, TX 77004, USA
| | - Wei Wang
- Bioinformatics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Ákos Végvári
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17 164, Solna, Sweden
| | - Isabella Salas-Allende
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | - Zakary Plautz
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | - Angel Cedazo-Minguez
- Department of Neurobiology Care Sciences and Society, Karolinska Institutet, 17 164, Solna, Sweden
| | - Subhash C Sinha
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Olga Troyanskaya
- Department of Computer Science, Princeton University, Princeton, NJ 08540, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY 10010, USA
| | - Marc Flajolet
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | - Vicky Yao
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17 164, Solna, Sweden
| | - Jean-Pierre Roussarie
- Department of Anatomy & Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
29
|
Nabizadeh F, Seyedmirzaei H, Karami S. Neuroimaging biomarkers and CSF sTREM2 levels in Alzheimer's disease: a longitudinal study. Sci Rep 2024; 14:15318. [PMID: 38961148 PMCID: PMC11222555 DOI: 10.1038/s41598-024-66211-w] [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: 10/27/2023] [Accepted: 06/28/2024] [Indexed: 07/05/2024] Open
Abstract
Understanding the exact pathophysiological mechanisms underlying the involvement of triggering receptor expressed on myeloid cells 2 (TREM2) related microglia activation is crucial for the development of clinical trials targeting microglia activation at different stages of Alzheimer's disease (AD). Given the contradictory findings in the literature, it is imperative to investigate the longitudinal alterations in cerebrospinal fluid (CSF) soluble TREM2 (sTREM2) levels as a marker for microglia activation, and its potential association with AD biomarkers, in order to address the current knowledge gap. In this study, we aimed to assess the longitudinal changes in CSF sTREM2 levels within the framework of the A/T/N classification system for AD biomarkers and to explore potential associations with AD pathological features, including the presence of amyloid-beta (Aβ) plaques and tau aggregates. The baseline and longitudinal (any available follow-up visit) CSF sTREM2 levels and processed tau-PET and Aβ-PET data of 1001 subjects were recruited from the ADNI database. The participants were classified into four groups based on the A/T/N framework: A+ /TN+ , A+ /TN- , A- /TN+ , and A- /TN- . Linear regression analyses were conducted to assess the relationship between CSF sTREM2 with cognitive performance, tau and Aβ-PET adjusting for age, gender, education, and APOE ε4 status. Based on our analysis there was a significant difference in baseline and rate of change of CSF sTREM2 between ATN groups. While there was no association between baseline CSF sTREM2 and cognitive performance (ADNI-mem), we found that the rate of change of CSF sTREM2 is significantly associated with cognitive performance in the entire cohort but not the ATN groups. We found that the baseline CSF sTREM2 is significantly associated with baseline tau-PET and Aβ-PET rate of change only in the A+ /TN+ group. A significant association was found between the rate of change of CSF sTREM2 and the tau- and Aβ-PET rate of change only in the A+ /TN- group. Our study suggests that the TREM2-related microglia activation and their relations with AD markers and cognitive performance vary the in presence or absence of Aβ and tau pathology. Furthermore, our findings revealed that a faster increase in the level of CSF sTREM2 might attenuate future Aβ plaque formation and tau aggregate accumulation only in the presence of Aβ pathology.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Alzheimer's Disease Institute, Tehran, Iran.
| | - Homa Seyedmirzaei
- School of Medicine, Tehran University of Medical Science, Tehran, Iran
- Interdisciplinary Neuroscience Research Program (INRP), Tehran University of Medical Sciences, Tehran, Iran
| | - Shaghayegh Karami
- School of Medicine, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
30
|
Wagatsuma K, Miwa K, Yamao T, Kamitaka Y, Akamatsu G, Nakajima K, Miyaji N, Ishibashi K, Ishii K. Development of a novel phantom for tau PET imaging. Phys Med 2024; 123:103399. [PMID: 38852366 DOI: 10.1016/j.ejmp.2024.103399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 06/02/2024] [Accepted: 06/05/2024] [Indexed: 06/11/2024] Open
Abstract
PURPOSE The cortical uptake of tau positron emission tomography (PET) tracers corresponds to the Braak stage and reflects the distribution and progression of tau neurofibrillary tangles. The present study aimed to develop and validate the basic performance of a novel tau PET phantom, as well as to establish standard test procedures and analytical methods. METHODS The tau PET phantom consisted of a brain simulation section simulated medial temporal lobe region and resolution and uniformity sections. The brain simulation section and hot rods and uniformity section contained 4 and 2 kBq/mL of 18F, respectively and images were acquired three times for 20 min with a PET/CT scanner. The resolution section was visually assessed with two sets of hot and cold rods. Recovery coefficients (RCs) as a quantitative value and coefficient of variation (CV) as image noise were determined based on the brain simulation and the uniformity section, respectively. RESULTS Preparation of activity in the phantom was repeatable among three measurements. The quality of images in the brain simulation and uniformity section with the rods was good. The 5- or 6-mm rods were detected separately. The mean RCs calculated based on the VOI template were between 0.75 and 0.83. The CV at the center slice of uniformity section was 5.54%. CONCLUSIONS We developed a novel tau PET phantom to assess quantitative value, image noise, and detectability and resolution from brain simulation section, uniformity section, and rods, respectively. This phantom will contribute to the standardization and harmonization of tau PET imaging.
Collapse
Affiliation(s)
- Kei Wagatsuma
- School of Allied Health Sciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan; Research Team for Neuroimaging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2, Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan.
| | - Kenta Miwa
- Department of Radiological Sciences, School of Health Sciences, Fukushima Medical University, 10-6 Sakaemachi, Fukushima-shi, Fukushima 960-8516, Japan
| | - Tensho Yamao
- Department of Radiological Sciences, School of Health Sciences, Fukushima Medical University, 10-6 Sakaemachi, Fukushima-shi, Fukushima 960-8516, Japan
| | - Yuto Kamitaka
- Research Team for Neuroimaging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2, Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan
| | - Go Akamatsu
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Kanta Nakajima
- School of Allied Health Sciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Noriaki Miyaji
- Department of Radiological Sciences, School of Health Sciences, Fukushima Medical University, 10-6 Sakaemachi, Fukushima-shi, Fukushima 960-8516, Japan
| | - Kenji Ishibashi
- Research Team for Neuroimaging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2, Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan
| | - Kenji Ishii
- Research Team for Neuroimaging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2, Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan
| |
Collapse
|
31
|
Stankeviciute L, Chhatwal JP, Levin R, Pinilla V, Schultz AP, Redline S, Johnson KA, Sperling RA, Kozhemiako N, Purcell S, Djonlagic I. Amyloid beta-independent sleep markers associated with early regional tau burden and cortical thinning. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12616. [PMID: 39077684 PMCID: PMC11284643 DOI: 10.1002/dad2.12616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION Sleep is crucial for memory consolidation and the clearance of toxic proteins associated with Alzheimer's disease (AD). We examined the association between sleep characteristics and imaging biomarkers of early amyloid beta (Aβ) and tau pathology as well as neurodegeneration in brain regions known to be affected in the incipient stages of AD. METHODS Thirty-nine cognitively unimpaired (CU) participants of the Harvard Aging Brain Study underwent at-home polysomnography as well as tau positron emission tomography (flortaucipir-PET), amyloid PET (Pittsburgh compound B [PiB]-PET), and magnetic resonance imaging-derived assessment of cortical thickness (CT). RESULTS Increased N1 sleep was associated with a higher tau PET signal (β = 0.009, p = 0.001) and lower CT in the temporal composite region of interest (β = -0.017, p = 0.007). Decreased slow-wave sleep (SWS) was associated with higher tau burden in the temporal composite (β = -0.008, p = 0.005) and lower CT (β = 0.008, p = 0.002), even after controlling for global PiB-PET. DISCUSSION In CU older adults, lower SWS and higher N1 sleep were associated with higher tau burden and lower CT in brain regions associated with early tau deposition and vulnerable to AD-related neurodegeneration through mechanisms dissociable from amyloid deposition. Highlights We report the results of an observational study, which leveraged -a well-characterized cohort of healthy aging (Harvard Aging Brain Study) by adding in-home full polysomnograms.By adding at-home polysomnograms to this unique and deeply phenotyped cohort, we examined variations in sleep architecture that are associated with Alzheimer's disease (AD) pathologic changes.Our results confirmed the association of sleep changes with early tau and cortical neurodegenerative changes that were independent of amyloid.The results will be of importance in monitoring sleep-related variations in relation to the natural history of AD pathology and in designing sleep-focused clinical trials.
Collapse
Affiliation(s)
- Laura Stankeviciute
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
| | - Jasmeer P. Chhatwal
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Raina Levin
- Massachusetts General HospitalBostonMassachusettsUSA
| | | | - Aaron P. Schultz
- Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Susan Redline
- Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Keith A. Johnson
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Reisa A. Sperling
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Nataliia Kozhemiako
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Shaun Purcell
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Ina Djonlagic
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
- Beth Israel Deaconess Medical CenterBostonMassachusettsUSA
| |
Collapse
|
32
|
Younes K, Smith V, Johns E, Carlson ML, Winer J, He Z, Henderson VW, Greicius MD, Young CB, Mormino EC. Temporal tau asymmetry spectrum influences divergent behavior and language patterns in Alzheimer's disease. Brain Behav Immun 2024; 119:807-817. [PMID: 38710339 DOI: 10.1016/j.bbi.2024.05.002] [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: 11/02/2023] [Revised: 03/31/2024] [Accepted: 05/02/2024] [Indexed: 05/08/2024] Open
Abstract
Understanding the psychiatric symptoms of Alzheimer s disease (AD) is crucial for advancing precision medicine and therapeutic strategies. The relationship between AD behavioral symptoms and asymmetry in spatial tau PET patterns is not well-known. Braak tau progression implicates the temporal lobes early. However, the clinical and pathological implications of temporal tau laterality remain unexplored. This cross-sectional study investigated the correlation between temporal tau PET asymmetry and behavior assessed using the neuropsychiatric inventory and composite scores for memory, executive function, and language, using data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) dataset. In the entire cohort, continuous right and left temporal tau contributions to behavior and cognition were evaluated, controlling for age, sex, education, and tau burden on the contralateral side. Additionally, a temporal tau laterality index was calculated to define "asymmetry-extreme" groups (individuals with laterality indices greater than two standard deviations from the mean). 695 individuals (age = 73.9 ± 7.6 years, 372 (53.5 %) females) were included, comprising 281 (40%) cognitively unimpaired (CU) amyloid negative, 185 (27%) CU amyloid positive, and 229 (33%) impaired (CI) amyloid positive participants. In the full cohort analysis, right temporal tau was associated with worse behavior (B = 8.14, p-value = 0.007), and left temporal tau was associated with worse language (B = 1.4, p-value < 0.001). Categorization into asymmetry-extreme groups revealed 20 right- and 27 left-asymmetric participants. Within these extreme groups, there was additional heterogeneity along the anterior-posterior dimension. Asymmetrical tau burden is associated with distinct behavioral and cognitive profiles. Wide multi-cultural implementation of social cognition measures is needed to understand right-sided asymmetry in AD.
Collapse
Affiliation(s)
- Kyan Younes
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, USA.
| | - Viktorija Smith
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, USA
| | - Emily Johns
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, USA
| | - Mackenzie L Carlson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, USA
| | - Joseph Winer
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, USA
| | - Zihuai He
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, USA; Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Victor W Henderson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, USA; Department of Epidemiology and Population Health, Stanford University, USA
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, USA
| | - Christina B Young
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, USA
| | - Elizabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, USA; Wu Tsai Neuroscience Institute, Stanford, CA, USA
| |
Collapse
|
33
|
Li J, Huang Q, Qi N, He K, Li S, Huang L, Pan F, Ren S, Hua F, Huang Y, Guan Y, Guo Q, Zhao J, Xie F. The associations between synaptic density and "A/T/N" biomarkers in Alzheimer's disease: An 18F-SynVesT-1 PET/MR study. J Cereb Blood Flow Metab 2024; 44:1199-1207. [PMID: 38295871 PMCID: PMC11179616 DOI: 10.1177/0271678x241230733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 12/08/2023] [Accepted: 12/26/2023] [Indexed: 06/13/2024]
Abstract
A newly developed SV2A radiotracer, 18F-SynVesT-1, was used in this study to investigate synaptic density and its association with Alzheimer's disease (AD) "A/T/N" biomarkers. The study included a cohort of 97 subjects, consisting of 64 patients with cognitive impairment (CI) and 33 individuals with normal cognition (CU). All subjects underwent 18F-SynVesT-1 PET/MR and 18F-florbetapir PET/CT scans. Additionally, a subgroup of individuals also underwent 18F-MK-6240, 18F-FDG PET/CT, plasma Aβ42/Aβ40 and p-tau181 tests. The differences in synaptic density between the groups and the correlations between synaptic density and AD "A/T/N" biomarkers were analyzed. The results showed that compared to the CU group, the CI with Aβ+ (CI+) group exhibited the most pronounced synapse loss in the hippocampus, with some loss also observed in the neocortex. Furthermore, synaptic density in the hippocampus and parahippocampal gyrus showed associations with AD biomarkers detected by both imaging and plasma tests in the CI group. The associations between synaptic density and FDG uptake and hippocampal volume were also observed in the CI+ group. In conclusion, the study demonstrated significant synaptic density loss, as measured by the promising tracer 18F-SynVesT-1, and its close correlation with "A/T/N" biomarkers in patients with both Alzheimer's clinical syndrome and pathological changes.
Collapse
Affiliation(s)
- Junpeng Li
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Huang
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Na Qi
- Department of Nuclear Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kun He
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Songye Li
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lin Huang
- Department of Gerontology, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Fengfeng Pan
- Department of Gerontology, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Shuhua Ren
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Fengchun Hua
- Department of Nuclear Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qihao Guo
- Department of Gerontology, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Jun Zhao
- Department of Nuclear Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
34
|
Strobel J, Yousefzadeh-Nowshahr E, Deininger K, Bohn KP, von Arnim CAF, Otto M, Solbach C, Anderl-Straub S, Polivka D, Fissler P, Glatting G, Riepe MW, Higuchi M, Beer AJ, Ludolph A, Winter G. Exploratory Tau PET/CT with [11C]PBB3 in Patients with Suspected Alzheimer's Disease and Frontotemporal Lobar Degeneration: A Pilot Study on Correlation with PET Imaging and Cerebrospinal Fluid Biomarkers. Biomedicines 2024; 12:1460. [PMID: 39062033 PMCID: PMC11274645 DOI: 10.3390/biomedicines12071460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/13/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Accurately diagnosing Alzheimer's disease (AD) and frontotemporal lobar degeneration (FTLD) is challenging due to overlapping symptoms and limitations of current imaging methods. This study investigates the use of [11C]PBB3 PET/CT imaging to visualize tau pathology and improve diagnostic accuracy. Given diagnostic challenges with symptoms and conventional imaging, [11C]PBB3 PET/CT's potential to enhance accuracy was investigated by correlating tau pathology with cerebrospinal fluid (CSF) biomarkers, positron emission tomography (PET), computed tomography (CT), amyloid-beta, and Mini-Mental State Examination (MMSE). We conducted [11C]PBB3 PET/CT imaging on 24 patients with suspected AD or FTLD, alongside [11C]PiB PET/CT (13 patients) and [18F]FDG PET/CT (15 patients). Visual and quantitative assessments of [11C]PBB3 uptake using standardized uptake value ratios (SUV-Rs) and correlation analyses with clinical assessments were performed. The scans revealed distinct tau accumulation patterns; 13 patients had no or faint uptake (PBB3-negative) and 11 had moderate to pronounced uptake (PBB3-positive). Significant inverse correlations were found between [11C]PBB3 SUV-Rs and MMSE scores, but not with CSF-tau or CSF-amyloid-beta levels. Here, we show that [11C]PBB3 PET/CT imaging can reveal distinct tau accumulation patterns and correlate these with cognitive impairment in neurodegenerative diseases. Our study demonstrates the potential of [11C]PBB3-PET imaging for visualizing tau pathology and assessing disease severity, offering a promising tool for enhancing diagnostic accuracy in AD and FTLD. Further research is essential to validate these findings and refine the use of tau-specific PET imaging in clinical practice, ultimately improving patient care and treatment outcomes.
Collapse
Affiliation(s)
- Joachim Strobel
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Katharina Deininger
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Karl Peter Bohn
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Markus Otto
- Department of Neurology, Halle University, 06120 Halle, Germany
| | - Christoph Solbach
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Dörte Polivka
- Department of Neurology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Patrick Fissler
- Psychiatric Services Thurgau (Academic Teaching Hospital of the University of Konstanz), 8596 Münsterlingen, Switzerland
| | - Gerhard Glatting
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Matthias W. Riepe
- Department of Psychiatry and Psychotherapy II, Ulm University, 89075 Ulm, Germany
| | - Makoto Higuchi
- National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Ambros J. Beer
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Albert Ludolph
- Department of Neurology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Gordon Winter
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| |
Collapse
|
35
|
Matthews DC, Kinney JW, Ritter A, Andrews RD, Toledano Strom EN, Lukic AS, Koenig LN, Revta C, Fillit HM, Zhong K, Tousi B, Leverenz JB, Feldman HH, Cummings J. Relationships between plasma biomarkers, tau PET, FDG PET, and volumetric MRI in mild to moderate Alzheimer's disease patients. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e12490. [PMID: 38988416 PMCID: PMC11233274 DOI: 10.1002/trc2.12490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/24/2024] [Accepted: 05/25/2024] [Indexed: 07/12/2024]
Abstract
INTRODUCTION The "A/T/N" (amyloid/tau/neurodegeneration) framework provides a biological basis for Alzheimer's disease (AD) diagnosis and can encompass additional changes such as inflammation ("I"). A spectrum of T/N/I imaging and plasma biomarkers was acquired in a phase 2 clinical trial of rasagiline in mild to moderate AD patients. We evaluated these to understand biomarker distributions and relationships within this population. METHODS Plasma biomarkers of pTau-181, neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), other inflammation-related proteins, imaging measures including fluorodeoxyglucose (FDG) positron emission tomography (PET), flortaucipir PET, and volumetric magnetic resonance imaging (MRI), and cognitive endpoints were analyzed to assess characteristics and relationships for the overall population (N = 47 at baseline and N = 21 for longitudinal cognitive comparisons) and within age-decade subgroups (57-69, 70-79, 80-90 years). RESULTS Data demonstrate wide clinical and biomarker heterogeneity in this population influenced by age and sex. Plasma pTau-181 and GFAP correlate with tau PET, most strongly in left inferior temporal cortex (p = 0.0002, p = 0.0006, respectively). In regions beyond temporal cortex, tau PET uptake decreased with age for the same pTau-181 or GFAP concentrations. FDG PET and brain volumes correlate with tau PET in numerous regions (such as inferior temporal: p = 0.0007, p = 0.00001, respectively). NfL, GFAP, and all imaging modalities correlate with baseline MMSE; subsequent MMSE decline is predicted by baseline parahippocampal and lateral temporal tau PET (p = 0.0007) and volume (p = 0.0006). Lateral temporal FDG PET (p = 0.006) and volume (p = 0.0001) are most strongly associated with subsequent ADAS-cog decline. NfL correlates with FDG PET and baseline MMSE but not tau PET. Inflammation biomarkers are intercorrelated but correlated with other biomarkers in only the youngest group. DISCUSSION Associations between plasma biomarkers, imaging biomarkers, and cognitive status observed in this study provide insight into relationships among biological processes in mild to moderate AD. Findings show the potential to characterize AD patients regarding likely tau pathology, neurodegeneration, prospective clinical decline, and the importance of covariates such as age. Highlights Plasma pTau-181 and GFAP correlated with regional and global tau PET in mild to moderate AD.NfL correlated with FDG PET and cognitive endpoints but not plasma pTau-181 or tau PET.Volume and FDG PET showed strong relationships to tau PET, one another, and cognitive status.Temporal volumes most strongly predicted decline in both MMSE and ADAS-cog.Volume and plasma biomarkers can enrich for elevated tau PET with age a significant covariate.
Collapse
Affiliation(s)
| | | | - Aaron Ritter
- Hoag Pickup Family Neurosciences InstituteNewport BeachCaliforniaUSA
| | | | - Erin N. Toledano Strom
- Chambers‐Grundy Center for Transformative NeuroscienceDepartment of Brain HealthSchool of Integrated Health Sciences, University of Nevada Las VegasLas VegasNevadaUSA
| | | | | | - Carolyn Revta
- Alzheimer's Disease Cooperative StudyUniversity of California, San Diego, School of MedicineLa JollaCaliforniaUSA
| | | | | | - Babak Tousi
- Cleveland Clinical Lous Ruvo Center for Brain HealthClevelandOhioUSA
| | | | - Howard H. Feldman
- Alzheimer's Disease Cooperative StudyUniversity of California, San Diego, School of MedicineLa JollaCaliforniaUSA
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Jeffrey Cummings
- Chambers‐Grundy Center for Transformative NeuroscienceDepartment of Brain HealthSchool of Integrated Health Sciences, University of Nevada Las VegasLas VegasNevadaUSA
| |
Collapse
|
36
|
Li K, Wang S, Luo X, Zeng Q, Liu X, Hong L, Li J, Hong H, Xu X, Zhang Y, Jiaerken Y, Zhang R, Xie L, Xu S, Zhang X, Chen Y, Liu Z, Zhang M, Huang P. Associations of Alzheimer's Disease Pathology and Small Vessel Disease With Cerebral White Matter Degeneration: A Tract-Based MR Diffusion Imaging Study. J Magn Reson Imaging 2024; 60:268-278. [PMID: 37737474 DOI: 10.1002/jmri.29022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 09/02/2023] [Accepted: 09/05/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND White matter (WM) degeneration is a key feature of Alzheimer's disease (AD). However, the underlying mechanism remains unclear. PURPOSE To investigate how amyloid-β (Aβ), tau, and small vascular disease (SVD) jointly affect WM degeneration in subjects along AD continuum. STUDY TYPE Retrospective. SUBJECTS 152 non-demented participants (age: 55.8-91.6, male/female: 66/86) from the ADNI database were included, classified into three groups using the A (Aβ)/T (tau)/N pathological scheme (Group 1: A-T-; Group 2: A+T-; Group 3: A+T+) based on positron emission tomography data. FIELD STRENGTH/SEQUENCE 3T; T1-weighted images, T2-weighted fluid-attenuated inversion recovery images, T2*-weighted images, diffusion-weighted spin-echo echo-planar imaging sequence (54 diffusion directions). ASSESSMENT Free-water diffusion model (generated parameters: free water, FW; tissue fractional anisotropy, FAt; tissue mean diffusivity, MDt); SVD total score; Neuropsychological tests. STATISTICAL TESTS Linear regression analysis was performed to investigate the independent contribution of AD (Aβ and tau) and SVD pathologies to diffusion parameters in each fiber tract, first in the entire population and then in each subgroup. We also investigated associations between diffusion parameters and cognitive functions. The level of statistical significance was set at p < 0.05 (false discovery rate corrected). RESULTS In the entire population, we found that: 1) Increased FW was significantly associated with SVD and tau, while FAt and MDt were significantly associated with Aβ and tau; 2) The spatial pattern of fiber tracts related to a certain pathological marker is consistent with the known distribution of that pathology; 3) Subgroup analysis showed that Group 2 and 3 had more alterations of FAt and MDt associated with Aβ and tau; 4) Diffusion imaging indices showed significant associations with cognitive score in all domains except memory. DATA CONCLUSION WM microstructural injury was associated with both AD and SVD pathologies, showing compartment-specific, tract-specific, and stage-specific WM patterns. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Kaicheng Li
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shuyue Wang
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Luo
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Qingze Zeng
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaocao Liu
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Luwei Hong
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jixuan Li
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Hong
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaopei Xu
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Zhang
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yeerfan Jiaerken
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Ruiting Zhang
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Linyun Xie
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shan Xu
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyi Zhang
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yanxing Chen
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zhirong Liu
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Peiyu Huang
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
37
|
Lee S, Kim SE, Jang H, Kim JP, Sohn G, Park YH, Ham H, Gu Y, Park CJ, Kim HJ, Na DL, Kim K, Seo SW. Distinct effects of blood pressure parameters on Alzheimer's and vascular markers in 1,952 Asian individuals without dementia. Alzheimers Res Ther 2024; 16:125. [PMID: 38863019 PMCID: PMC11167921 DOI: 10.1186/s13195-024-01483-y] [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: 11/12/2023] [Accepted: 05/15/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND Risk factors for cardiovascular disease, including elevated blood pressure, are known to increase risk of Alzheimer's disease. There has been increasing awareness of the relationship between long-term blood pressure (BP) patterns and their effects on the brain. We aimed to investigate the association of repeated BP measurements with Alzheimer's and vascular disease markers. METHODS We recruited 1,952 participants without dementia between August 2015 and February 2022. During serial clinic visits, we assessed both systolic BP (SBP) and diastolic BP (DBP), and visit-to-visit BP variability (BPV) was quantified from repeated measurements. In order to investigate the relationship of mean SBP (or DBP) with Alzheimer's and vascular markers and cognition, we performed multiple linear and logistic regression analyses after controlling for potential confounders (Model 1). Next, we investigated the relationship of with variation of SBP (or DBP) with the aforementioned variables by adding it into Model 1 (Model 2). In addition, mediation analyses were conducted to determine mediation effects of Alzheimer's and vascular makers on the relationship between BP parameters and cognitive impairment. RESULTS High Aβ uptake was associated with greater mean SBP (β = 1.049, 95% confidence interval 1.016-1.083). High vascular burden was positively associated with mean SBP (odds ratio = 1.293, 95% CI 1.015-1.647) and mean DBP (1.390, 1.098-1.757). High tau uptake was related to greater systolic BPV (0.094, 0.001-0.187) and diastolic BPV (0.096, 0.007-0.184). High Aβ uptake partially mediated the relationship between mean SBP and the Mini-Mental State Examination (MMSE) scores. Hippocampal atrophy mediated the relationship between diastolic BPV and MMSE scores. CONCLUSIONS Each BP parameter affects Alzheimer's and vascular disease markers differently, which in turn leads to cognitive impairment. Therefore, it is necessary to appropriately control specific BP parameters to prevent the development of dementia. Furthermore, a better understanding of pathways from specific BP parameters to cognitive impairments might enable us to select the managements targeting the specific BP parameters to prevent dementia effectively.
Collapse
Affiliation(s)
- Sungjoo Lee
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Si Eun Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Department of Neurology, Inje University College of Medicine, Haeundae Paik Hospital, Busan, 48108, Republic of Korea
| | - Hyemin Jang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jun Pyo Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Gyeongmo Sohn
- Department of Neurology, Inje University College of Medicine, Haeundae Paik Hospital, Busan, 48108, Republic of Korea
| | - Yu Hyun Park
- Neuroscience Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Hongki Ham
- Neuroscience Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Yuna Gu
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Chae Jung Park
- Research Institute, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Duk L Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Kyunga Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- Biomedical Statistics Center, Research Institute for Future Medicine, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- Department of Data Convergence & Future Medicine, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- Neuroscience Center, Samsung Medical Center, Seoul, 06351, Republic of Korea.
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, 06351, Republic of Korea.
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Republic of Korea.
- Center for Clinical Epidemiology, Samsung Medical Center, Seoul, 06351, Republic of Korea.
- Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul, 06351, Republic of Korea.
| |
Collapse
|
38
|
Denkinger M, Baker S, Inglis B, Kobayashi S, Juarez A, Mason S, Jagust W. Associations between regional blood-brain barrier permeability, aging, and Alzheimer's disease biomarkers in cognitively normal older adults. PLoS One 2024; 19:e0299764. [PMID: 38837947 PMCID: PMC11152304 DOI: 10.1371/journal.pone.0299764] [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: 02/20/2024] [Accepted: 05/05/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Increased blood-brain barrier permeability (BBBp) has been hypothesized as a feature of aging that may lead to the development of Alzheimer's disease (AD). We sought to identify the brain regions most vulnerable to greater BBBp during aging and examine their regional relationship with neuroimaging biomarkers of AD. METHODS We studied 31 cognitively normal older adults (OA) and 10 young adults (YA) from the Berkeley Aging Cohort Study (BACS). Both OA and YA received dynamic contrast-enhanced MRI (DCE-MRI) to quantify Ktrans values, as a measure of BBBp, in 37 brain regions across the cortex. The OA also received Pittsburgh compound B (PiB)-PET to create distribution volume ratios (DVR) images and flortaucipir (FTP)- PET to create partial volume corrected standardized uptake volume ratios (SUVR) images. Repeated measures ANOVA assessed the brain regions where OA showed greater BBBp than YA. In OA, Ktrans values were compared based on sex, Aβ positivity status, and APOE4 carrier status within a composite region across the areas susceptible to aging. We used linear models and sparse canonical correlation analysis (SCCA) to examine the relationship between Ktrans and AD biomarkers. RESULTS OA showed greater BBBp than YA predominately in the temporal lobe, with some involvement of parietal, occipital and frontal lobes. Within an averaged ROI of affected regions, there was no difference in Ktrans values based on sex or Aβ positivity, but OA who were APOE4 carriers had significantly higher Ktrans values. There was no direct relationship between averaged Ktrans and global Aβ pathology, but there was a trend for an Ab status by tau interaction on Ktrans in this region. SCCA showed increased Ktrans was associated with increased PiB DVR, mainly in temporal and parietal brain regions. There was not a significant relationship between Ktrans and FTP SUVR. DISCUSSION Our findings indicate that the BBB shows regional vulnerability during normal aging that overlaps considerably with the pattern of AD pathology. Greater BBBp in brain regions affected in aging is related to APOE genotype and may also be related to the pathological accumulation of Aβ.
Collapse
Affiliation(s)
- Marisa Denkinger
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, United States of America
| | - Suzanne Baker
- Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Ben Inglis
- Henry H. Wheeler Jr. Brain Imaging Center, University of California, Berkeley, Berkeley, California, United States of America
| | - Sarah Kobayashi
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, United States of America
| | - Alexis Juarez
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, United States of America
| | - Suzanne Mason
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, United States of America
| | - William Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, United States of America
- Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| |
Collapse
|
39
|
Cody KA, Langhough RE, Zammit MD, Clark L, Chin N, Christian BT, Betthauser TJ, Johnson SC. Characterizing brain tau and cognitive decline along the amyloid timeline in Alzheimer's disease. Brain 2024; 147:2144-2157. [PMID: 38667631 PMCID: PMC11146417 DOI: 10.1093/brain/awae116] [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: 10/30/2023] [Revised: 02/23/2024] [Accepted: 03/24/2024] [Indexed: 06/04/2024] Open
Abstract
Recent longitudinal PET imaging studies have established methods to estimate the age at which amyloid becomes abnormal at the level of the individual. Here we recontextualized amyloid levels into the temporal domain to better understand the downstream Alzheimer's disease processes of tau neurofibrillary tangle (NFT) accumulation and cognitive decline. This cohort study included a total of 601 individuals from the Wisconsin Registry for Alzheimer's Prevention and Wisconsin Alzheimer's Disease Research Center that underwent amyloid and tau PET, longitudinal neuropsychological assessments and met clinical criteria for three clinical diagnosis groups: cognitively unimpaired (n = 537); mild cognitive impairment (n = 48); or dementia (n = 16). Cortical 11C-Pittsburgh compound B (PiB) distribution volume ratio (DVR) and sampled iterative local approximation were used to estimate amyloid positive (A+; global PiB DVR > 1.16 equivalent to 17.1 centiloids) onset age and years of A+ duration at tau PET (i.e. amyloid chronicity). Tau PET burden was quantified using 18F-MK-6240 standardized uptake value ratios (70-90 min, inferior cerebellar grey matter reference region). Whole-brain and region-specific approaches were used to examine tau PET binding along the amyloid timeline and across the Alzheimer's disease clinical continuum. Voxel-wise 18F-MK-6240 analyses revealed that with each decade of A+, the spatial extent of measurable tau spread (i.e. progressed) from regions associated with early to late NFT tau stages. Regional analyses indicated that tau burden in the entorhinal cortex was detectable, on average, within 10 years of A+ onset. Additionally, the entorhinal cortex was the region most sensitive to early amyloid pathology and clinical impairment in this predominantly preclinical sample. Among initially cognitively unimpaired (n = 472) individuals with longitudinal cognitive follow-up, mixed effects models showed significant linear and non-linear interactions of A+ duration and entorhinal tau on cognitive decline, suggesting a synergistic effect whereby greater A+ duration, together with a higher entorhinal tau burden, increases the likelihood of cognitive decline beyond their separable effects. Overall, the amyloid time framework enabled a spatiotemporal characterization of tau deposition patterns across the Alzheimer's disease continuum. This approach, which examined cross-sectional tau PET data along the amyloid timeline to make longitudinal disease course inferences, demonstrated that A+ duration explains a considerable amount of variability in the magnitude and topography of tau spread, which largely recapitulated NFT staging observed in human neuropathological studies. By anchoring disease progression to the onset of amyloid, this study provides a temporal disease context, which may help inform disease prognosis and timing windows for anti-amyloid therapies.
Collapse
Affiliation(s)
- Karly A Cody
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Rebecca E Langhough
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Matthew D Zammit
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, WI 53792, USA
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Lindsay Clark
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Nathaniel Chin
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Bradley T Christian
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, WI 53792, USA
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Tobey J Betthauser
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| |
Collapse
|
40
|
Feng Y, Laraib A, Lin X, Li Q, Zhan J, Li X. Associations of tau, Aβ, and brain volume of the Papez circuit with cognition in Alzheimer's disease. Eur Arch Psychiatry Clin Neurosci 2024:10.1007/s00406-024-01827-7. [PMID: 38824476 DOI: 10.1007/s00406-024-01827-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 05/10/2024] [Indexed: 06/03/2024]
Abstract
This study aimed to investigate the cross-sectional associations between regional Alzheimer's disease (AD) biomarkers, including tau, β-amyloid (Aβ), and brain volume, within the Papez circuit, and neuropsychological functioning across the preclinical and clinical spectrum of AD. We utilized data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database, including 251 Aβ-positive participants. Participants were categorized into three groups based on the Clinical Dementia Rating (CDR): 73 individuals with preclinical AD (CDR = 0), 114 with prodromal AD (CDR = 0.5), and 64 with clinical AD dementia (CDR ≥ 1). Linear regression analyses, adjusted for age, gender, and education years, were employed to evaluate the associations between five regions of interest (the hippocampus, para-hippocampus, entorhinal cortex, posterior cingulate cortex, and thalamus) and five neuropsychological tests across the three imaging modalities. In the preclinical stage of AD, flortaucipir PET was associated with impaired global cognition and episodic memory (range standardized β = 0.255-0.498, p < 0.05 corrected for multiple comparisons), while florbetapir PET and brain volume were marginally related to global cognition (range standardized β = 0.221-0.231, p < 0.05). In the clinical stages of AD (prodromal and dementia), both increased flortaucipir uptake and decreased brain volume were significantly associated with poorer global neuropsychological and episodic memory performance (range standardized β = 0.222-0.621, p < 0.05, most regions of interest survived correction for multiple comparisions). However, a slight relationship was observed between florbetapir uptake and poorer global cognitive function. The regions most affected by flortaucipir PET were the hippocampus, para-hippocampus, and posterior cingulate cortex. During the clinical stages, the hippocampus and entorhinal cortex exhibited the most significant volumetric changes. Tau PET and brain volume measurements within the Papez circuit are more sensitive indicators of early cognitive deficits in AD than Aβ PET. Furthermore, during the clinical stages of AD, both flortaucipir PET and brain volume of the Papez circuit are closely correlated with cognitive decline. These findings underscore the importance of integrating multiple biomarkers for the comprehensive evaluation of AD pathology and its impact on cognition.
Collapse
Affiliation(s)
- Yuxue Feng
- Department of Neurology, The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Azka Laraib
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74-76 Linjiang Road, Yuzhong District, Chongqing, 400000, China
| | - Xiuqi Lin
- Chongqing Medical University, Chongqing, China
| | - Qin Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74-76 Linjiang Road, Yuzhong District, Chongqing, 400000, China
| | - Jiehong Zhan
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74-76 Linjiang Road, Yuzhong District, Chongqing, 400000, China
| | - Xiaofeng Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74-76 Linjiang Road, Yuzhong District, Chongqing, 400000, China.
- Department of Neurology, People's Hospital of Linshui County, Guangan, China.
| |
Collapse
|
41
|
Earnest T, Bani A, Ha SM, Hobbs DA, Kothapalli D, Yang B, Lee JJ, Benzinger TLS, Gordon BA, Sotiras A. Data-driven decomposition and staging of flortaucipir uptake in Alzheimer's disease. Alzheimers Dement 2024; 20:4002-4019. [PMID: 38683905 PMCID: PMC11180875 DOI: 10.1002/alz.13769] [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: 10/10/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION Previous approaches pursuing in vivo staging of tau pathology in Alzheimer's disease (AD) have typically relied on neuropathologically defined criteria. In using predefined systems, these studies may miss spatial deposition patterns which are informative of disease progression. METHODS We selected discovery (n = 418) and replication (n = 132) cohorts with flortaucipir imaging. Non-negative matrix factorization (NMF) was applied to learn tau covariance patterns and develop a tau staging system. Flortaucipir components were also validated by comparison with amyloid burden, gray matter loss, and the expression of AD-related genes. RESULTS We found eight flortaucipir covariance patterns which were reproducible and overlapped with relevant gene expression maps. Tau stages were associated with AD severity as indexed by dementia status and neuropsychological performance. Comparisons of flortaucipir uptake with amyloid and atrophy also supported our model of tau progression. DISCUSSION Data-driven decomposition of flortaucipir uptake provides a novel framework for tau staging which complements existing systems. HIGHLIGHTS NMF reveals patterns of tau deposition in AD. Data-driven staging of flortaucipir tracks AD severity. Learned flortaucipir patterns overlap with AD-related gene expression.
Collapse
Affiliation(s)
- Tom Earnest
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Abdalla Bani
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Sung Min Ha
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Diana A. Hobbs
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Deydeep Kothapalli
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Braden Yang
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - John J. Lee
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Tammie L. S. Benzinger
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Brian A. Gordon
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Aristeidis Sotiras
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
- Institute for Informatics, Data Science & BiostatisticsWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | | |
Collapse
|
42
|
Shapiro ALB, Coughlan C, Bettcher BM, Pauley ME, Kim J, Bjornstad P, Rajic B, Truong J, Bell C, Choi YJ, Walker KA, Potter H, Liese AD, Dabelea D, Whitlow CT. Biomarkers of Neurodegeneration and Alzheimer's Disease Neuropathology in Adolescents and Young Adults with Youth-Onset Type 1 or Type 2 Diabetes: A Proof-of-Concept Study. ENDOCRINES 2024; 5:197-213. [PMID: 38764894 PMCID: PMC11101213 DOI: 10.3390/endocrines5020014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2024] Open
Abstract
Adult-onset diabetes increases one's risk of neurodegenerative disease including Alzheimer's disease (AD); however, the risk associated with youth-onset diabetes (Y-DM) remains underexplored. We quantified plasma biomarkers of neurodegeneration and AD in participants with Y-DM from the SEARCH cohort at adolescence and young adulthood (Type 1, n = 25; Type 2, n = 25; 59% female; adolescence, age = 15 y/o [2.6]; adulthood, age = 27.4 y/o [2.2]), comparing them with controls (adolescence, n = 25, age = 14.8 y/o [2.7]; adulthood, n = 21, age = 24.9 y/o [2.8]). Plasma biomarkers, including glial fibrillary acidic protein (GFAP), neurofilament light chain protein (NfL), phosphorylated tau-181 (pTau181), and amyloid beta (Aβ40, Aβ42), were measured via Simoa. A subset of participants (n = 7; age = 27.5 y/o [5.7]) and six controls (age = 25.1 y/o [4.5]) underwent PET scans to quantify brain amyloid and tau densities in AD sensitive brain regions. Y-DM adolescents exhibited lower plasma levels of Aβ40, Aβ42, and GFAP, and higher pTau181 compared to controls (p < 0.05), a pattern persisting into adulthood (p < 0.001). All biomarkers showed significant increases from adolescence to adulthood in Y-DM (p < 0.01), though no significant differences in brain amyloid or tau were noted between Y-DM and controls in adulthood. Preliminary evidence suggests that preclinical AD neuropathology is present in young people with Y-DM, indicating a potential increased risk of neurodegenerative diseases.
Collapse
Affiliation(s)
- Allison L. B. Shapiro
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado at Anschutz (CU-Anschutz), Aurora, CO 80045, USA
- Section of Endocrinology, Department of Pediatrics, School of Medicine (SOM), CU-Anschutz, Aurora, CO 80045, USA
| | - Christina Coughlan
- University of Colorado Alzheimer’s and Cognition Center, CU-Anschutz, Aurora, CO 80045, USA
- Department of Neurology, SOM, CU-Anschutz, Aurora, CO 80045, USA
| | | | - Meghan E. Pauley
- Barbara Davis Center for Diabetes, CU-Anschutz, Aurora, CO 80045, USA
| | - Jeongchul Kim
- Radiology Informatics and Image Processing Laboratory, Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Petter Bjornstad
- Section of Endocrinology, Department of Pediatrics, School of Medicine (SOM), CU-Anschutz, Aurora, CO 80045, USA
- Division of Renal Diseases and Hypertension, Department of Medicine, SOM, CU-Anschutz, Aurora, CO 80045, USA
| | - Benjamin Rajic
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado at Anschutz (CU-Anschutz), Aurora, CO 80045, USA
| | - Jennifer Truong
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado at Anschutz (CU-Anschutz), Aurora, CO 80045, USA
| | - Christopher Bell
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado at Anschutz (CU-Anschutz), Aurora, CO 80045, USA
| | - Ye Ji Choi
- Section of Endocrinology, Department of Pediatrics, School of Medicine (SOM), CU-Anschutz, Aurora, CO 80045, USA
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD 20814, USA
| | - Huntington Potter
- University of Colorado Alzheimer’s and Cognition Center, CU-Anschutz, Aurora, CO 80045, USA
- Department of Neurology, SOM, CU-Anschutz, Aurora, CO 80045, USA
| | - Angela D. Liese
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Dana Dabelea
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado at Anschutz (CU-Anschutz), Aurora, CO 80045, USA
| | - Christopher T. Whitlow
- Radiology Informatics and Image Processing Laboratory, Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| |
Collapse
|
43
|
Karimani F, Asgari Taei A, Abolghasemi-Dehaghani MR, Safari MS, Dargahi L. Impairment of entorhinal cortex network activity in Alzheimer's disease. Front Aging Neurosci 2024; 16:1402573. [PMID: 38882526 PMCID: PMC11176617 DOI: 10.3389/fnagi.2024.1402573] [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: 03/17/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
The entorhinal cortex (EC) stands out as a critical brain region affected in the early phases of Alzheimer's disease (AD), with some of the disease's pathological processes originating from this area, making it one of the most crucial brain regions in AD. Recent research highlights disruptions in the brain's network activity, characterized by heightened excitability and irregular oscillations, may contribute to cognitive impairment. These disruptions are proposed not only as potential therapeutic targets but also as early biomarkers for AD. In this paper, we will begin with a review of the anatomy and function of EC, highlighting its selective vulnerability in AD. Subsequently, we will discuss the disruption of EC network activity, exploring changes in excitability and neuronal oscillations in this region during AD and hypothesize that, considering the advancements in neuromodulation techniques, addressing the disturbances in the network activity of the EC could offer fresh insights for both the diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Farnaz Karimani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afsaneh Asgari Taei
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mir-Shahram Safari
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
Mathoux G, Boccalini C, Peretti DE, Arnone A, Ribaldi F, Scheffler M, Frisoni GB, Garibotto V. A comparison of visual assessment and semi-quantification for the diagnostic and prognostic use of [ 18F]flortaucipir PET in a memory clinic cohort. Eur J Nucl Med Mol Imaging 2024; 51:1639-1650. [PMID: 38182839 DOI: 10.1007/s00259-023-06583-9] [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: 10/04/2023] [Accepted: 12/22/2023] [Indexed: 01/07/2024]
Abstract
PURPOSE [18F]Flortaucipir PET is a powerful diagnostic and prognostic tool for Alzheimer's disease (AD). Tau status definition is mainly based in the literature on semi-quantitative measures while in clinical settings visual assessment is usually preferred. We compared visual assessment with established semi-quantitative measures to classify subjects and predict the risk of cognitive decline in a memory clinic population. METHODS We included 245 individuals from the Geneva Memory Clinic who underwent [18F]flortaucipir PET. Amyloid status was available for 207 individuals and clinical follow-up for 135. All scans were blindly evaluated by three independent raters who visually classified the scans according to Braak stages. Standardized uptake value ratio (SUVR) values were obtained from a global meta-ROI to define tau positivity, and the Simplified Temporo-Occipital Classification (STOC) was applied to obtain semi-quantitatively tau stages. The agreement between measures was tested using Cohen's kappa (k). ROC analysis and linear mixed-effects models were applied to test the diagnostic and prognostic values of tau status and stages obtained with the visual and semi-quantitative approaches. RESULTS We found good inter-rater reliability in the visual interpretation of tau Braak stages, independently from the rater's expertise (k>0.68, p<0.01). A good agreement was equally found between visual and SUVR-based classifications for tau status (k=0.67, p<0.01). All tau-assessment modalities significantly discriminated amyloid-positive MCI and demented subjects from others (AUC>0.80) and amyloid-positive from negative subjects (AUC>0.85). Linear mixed-effect models showed that tau-positive individuals presented a significantly faster cognitive decline than the tau-negative group (p<0.01), independently from the classification method. CONCLUSION Our results show that visual assessment is reliable for defining tau status and stages in a memory clinic population. The high inter-rater reliability, the substantial agreement, and the similar diagnostic and prognostic performance of visual rating and semi-quantitative methods demonstrate that [18F]flortaucipir PET can be robustly assessed visually in clinical practice.
Collapse
Affiliation(s)
- Gregory Mathoux
- Diagnostic Department, Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
- Università degli Studi Milano-Bicocca, Milano, Italy
| | - Cecilia Boccalini
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Faculty of Medicine, Geneva University Neurocenter, University of Geneva , Geneva, Switzerland
- Università Vita e Salute San Raffaele, Milano, Italy
| | - Debora E Peretti
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Faculty of Medicine, Geneva University Neurocenter, University of Geneva , Geneva, Switzerland
| | - Annachiara Arnone
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Faculty of Medicine, Geneva University Neurocenter, University of Geneva , Geneva, Switzerland
| | - Federica Ribaldi
- Department of Rehabilitation and Geriatrics, Memory Clinic, Geneva University and University Hospitals, Geneva, Switzerland
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
| | - Max Scheffler
- Division of Radiology, Geneva University Hospitals, Geneva, Switzerland
| | - Giovanni B Frisoni
- Department of Rehabilitation and Geriatrics, Memory Clinic, Geneva University and University Hospitals, Geneva, Switzerland
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
| | - Valentina Garibotto
- Diagnostic Department, Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals, University of Geneva, Geneva, Switzerland.
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Faculty of Medicine, Geneva University Neurocenter, University of Geneva , Geneva, Switzerland.
- CIBM Center for Biomedical Imaging, Geneva, Switzerland.
| |
Collapse
|
45
|
Jiang J, Shi R, Lu J, Wang M, Zhang Q, Zhang S, Wang L, Alberts I, Rominger A, Zuo C, Shi K. Detection of individual brain tau deposition in Alzheimer's disease based on latent feature-enhanced generative adversarial network. Neuroimage 2024; 291:120593. [PMID: 38554780 DOI: 10.1016/j.neuroimage.2024.120593] [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: 11/27/2023] [Revised: 03/17/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024] Open
Abstract
OBJECTIVE The conventional methods for interpreting tau PET imaging in Alzheimer's disease (AD), including visual assessment and semi-quantitative analysis of fixed hallmark regions, are insensitive to detect individual small lesions because of the spatiotemporal neuropathology's heterogeneity. In this study, we proposed a latent feature-enhanced generative adversarial network model for the automatic extraction of individual brain tau deposition regions. METHODS The latent feature-enhanced generative adversarial network we propose can learn the distribution characteristics of tau PET images of cognitively normal individuals and output the abnormal distribution regions of patients. This model was trained and validated using 1131 tau PET images from multiple centres (with distinct races, i.e., Caucasian and Mongoloid) with different tau PET ligands. The overall quality of synthetic imaging was evaluated using structural similarity (SSIM), peak signal to noise ratio (PSNR), and mean square error (MSE). The model was compared to the fixed templates method for diagnosing and predicting AD. RESULTS The reconstructed images archived good quality, with SSIM = 0.967 ± 0.008, PSNR = 31.377 ± 3.633, and MSE = 0.0011 ± 0.0007 in the independent test set. The model showed higher classification accuracy (AUC = 0.843, 95 % CI = 0.796-0.890) and stronger correlation with clinical scales (r = 0.508, P < 0.0001). The model also achieved superior predictive performance in the survival analysis of cognitive decline, with a higher hazard ratio: 3.662, P < 0.001. INTERPRETATION The LFGAN4Tau model presents a promising new approach for more accurate detection of individualized tau deposition. Its robustness across tracers and races makes it a potentially reliable diagnostic tool for AD in practice.
Collapse
Affiliation(s)
- Jiehui Jiang
- Institute of Biomedical Engineering, School of Life Sciences, Shanghai University, Shanghai, China.
| | - Rong Shi
- School of Information and Communication Engineering, Shanghai University, Shanghai, China
| | - Jiaying Lu
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China; National Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Min Wang
- Institute of Biomedical Engineering, School of Life Sciences, Shanghai University, Shanghai, China.
| | - Qi Zhang
- School of Information and Communication Engineering, Shanghai University, Shanghai, China
| | - Shuoyan Zhang
- School of Information and Communication Engineering, Shanghai University, Shanghai, China
| | - Luyao Wang
- Institute of Biomedical Engineering, School of Life Sciences, Shanghai University, Shanghai, China
| | - Ian Alberts
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland
| | - Chuantao Zuo
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China; National Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China; Human Phenome Institute, Fudan University, Shanghai, China.
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland; Department of Informatics, Technical University of Munich, Munich, Germany
| |
Collapse
|
46
|
Gérard T, Colmant L, Malotaux V, Salman Y, Huyghe L, Quenon L, Dricot L, Ivanoiu A, Lhommel R, Hanseeuw B. The spatial extent of tauopathy on [ 18F]MK-6240 tau PET shows stronger association with cognitive performances than the standard uptake value ratio in Alzheimer's disease. Eur J Nucl Med Mol Imaging 2024; 51:1662-1674. [PMID: 38228971 PMCID: PMC11043108 DOI: 10.1007/s00259-024-06603-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/04/2024] [Indexed: 01/18/2024]
Abstract
PURPOSE [18F]MK-6240, a second-generation tau PET tracer, is increasingly used for the detection and the quantification of in vivo cerebral tauopathy in Alzheimer's disease (AD). Given that neurological symptoms are better explained by the topography rather than by the nature of brain lesions, our study aimed to evaluate whether cognitive impairment would be more closely associated with the spatial extent than with the intensity of tau-PET signal, as measured by the standard uptake value ratio (SUVr). METHODS [18F]MK6240 tau-PET data from 82 participants in the AD spectrum were quantified in three different brain regions (Braak ≤ 2, Braak ≤ 4, and Braak ≤ 6) using SUVr and the extent of tauopathy (EOT, percentage of voxels with SUVr ≥ 1.3). PET data were first compared between diagnostic categories, and ROC curves were computed to evaluate sensitivity and specificity. PET data were then correlated to cognitive performances and cerebrospinal fluid (CSF) tau values. RESULTS The EOT in the Braak ≤ 2 region provided the highest diagnostic accuracies, distinguishing between amyloid-negative and positive clinically unimpaired individuals (threshold = 9%, sensitivity = 79%, specificity = 82%) as well as between prodromal AD and preclinical AD (threshold = 38%, sensitivity = 81%, specificity = 93%). The EOT better correlated with cognition than SUVr (∆R2 + 0.08-0.09) with the best correlation observed for EOT in the Braak ≤ 4 region (R2 = 0.64). Cognitive performances were more closely associated with PET metrics than with CSF values. CONCLUSIONS Quantifying [18F]MK-6240 tau PET in terms of EOT rather than SUVr significantly increases the correlation with cognitive performances. Quantification in the mesiotemporal lobe is the most useful to diagnose preclinical AD or prodromal AD.
Collapse
Affiliation(s)
- Thomas Gérard
- Nuclear Medicine Department, Cliniques Universitaires Saint Luc, Brussels, Belgium.
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium.
| | - Lise Colmant
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
- Neurology Department, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Vincent Malotaux
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
| | - Yasmine Salman
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
| | - Lara Huyghe
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
| | - Lisa Quenon
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
- Neurology Department, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Laurence Dricot
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
| | - Adrian Ivanoiu
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
- Neurology Department, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Renaud Lhommel
- Nuclear Medicine Department, Cliniques Universitaires Saint Luc, Brussels, Belgium
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
| | - Bernard Hanseeuw
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
- Neurology Department, Cliniques Universitaires Saint Luc, Brussels, Belgium
- WELBIO Department, WEL Research Institute, Avenue Pasteur, 6, 1300, Wavre, Belgium
- Department of Radiology, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
47
|
Wang M, Lu J, Zhang Y, Zhang Q, Wang L, Wu P, Brendel M, Rominger A, Shi K, Zhao Q, Jiang J, Zuo C. Characterization of tau propagation pattern and cascading hypometabolism from functional connectivity in Alzheimer's disease. Hum Brain Mapp 2024; 45:e26689. [PMID: 38703095 PMCID: PMC11069321 DOI: 10.1002/hbm.26689] [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: 01/17/2024] [Revised: 03/16/2024] [Accepted: 04/08/2024] [Indexed: 05/06/2024] Open
Abstract
Tau pathology and its spatial propagation in Alzheimer's disease (AD) play crucial roles in the neurodegenerative cascade leading to dementia. However, the underlying mechanisms linking tau spreading to glucose metabolism remain elusive. To address this, we aimed to examine the association between pathologic tau aggregation, functional connectivity, and cascading glucose metabolism and further explore the underlying interplay mechanisms. In this prospective cohort study, we enrolled 79 participants with 18F-Florzolotau positron emission tomography (PET), 18F-fluorodeoxyglucose PET, resting-state functional, and anatomical magnetic resonance imaging (MRI) images in the hospital-based Shanghai Memory Study. We employed generalized linear regression and correlation analyses to assess the associations between Florzolotau accumulation, functional connectivity, and glucose metabolism in whole-brain and network-specific manners. Causal mediation analysis was used to evaluate whether functional connectivity mediates the association between pathologic tau and cascading glucose metabolism. We examined 22 normal controls and 57 patients with AD. In the AD group, functional connectivity was associated with Florzolotau covariance (β = .837, r = 0.472, p < .001) and glucose covariance (β = 1.01, r = 0.499, p < .001). Brain regions with higher tau accumulation tend to be connected to other regions with high tau accumulation through functional connectivity or metabolic connectivity. Mediation analyses further suggest that functional connectivity partially modulates the influence of tau accumulation on downstream glucose metabolism (mediation proportion: 49.9%). Pathologic tau may affect functionally connected neurons directly, triggering downstream glucose metabolism changes. This study sheds light on the intricate relationship between tau pathology, functional connectivity, and downstream glucose metabolism, providing critical insights into AD pathophysiology and potential therapeutic targets.
Collapse
Affiliation(s)
- Min Wang
- School of Life SciencesShanghai UniversityShanghaiChina
| | - Jiaying Lu
- Department of Nuclear Medicine & PET Center, Huashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and Medicine, Huashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological Disorders, Huashan HospitalFudan UniversityShanghaiChina
| | - Ying Zhang
- School of Life SciencesShanghai UniversityShanghaiChina
| | - Qi Zhang
- School of Life SciencesShanghai UniversityShanghaiChina
| | - Luyao Wang
- School of Life SciencesShanghai UniversityShanghaiChina
| | - Ping Wu
- Department of Nuclear Medicine & PET Center, Huashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and Medicine, Huashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological Disorders, Huashan HospitalFudan UniversityShanghaiChina
| | | | - Axel Rominger
- Department of Nuclear Medicine, InselspitalBern University Hospital, University of BernBernSwitzerland
| | - Kuangyu Shi
- Department of Nuclear Medicine, InselspitalBern University Hospital, University of BernBernSwitzerland
- Computer Aided Medical Procedures, School of Computation, Information and TechnologyTechnical University of MunichMunichGermany
| | - Qianhua Zhao
- National Clinical Research Center for Aging and Medicine, Huashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological Disorders, Huashan HospitalFudan UniversityShanghaiChina
- Department of Neurology, Huashan HospitalFudan UniversityShanghaiChina
| | - Jiehui Jiang
- School of Life SciencesShanghai UniversityShanghaiChina
| | - Chuantao Zuo
- Department of Nuclear Medicine & PET Center, Huashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and Medicine, Huashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological Disorders, Huashan HospitalFudan UniversityShanghaiChina
- Human Phenome InstituteFudan UniversityShanghaiChina
| |
Collapse
|
48
|
Gandia-Ferrero MT, Torres-Espallardo I, Martínez-Sanchis B, Muñoz E, Morera-Ballester C, Sopena-Novales P, Álvarez-Sánchez L, Baquero-Toledo M, Martí-Bonmatí L. Amyloid brain-dedicated PET images can diagnose Alzheimer's pathology with Centiloid Scale. Phys Med 2024; 121:103345. [PMID: 38581963 DOI: 10.1016/j.ejmp.2024.103345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 03/15/2024] [Accepted: 04/01/2024] [Indexed: 04/08/2024] Open
Abstract
PURPOSE To evaluate whether the Centiloid Scale may be used to diagnose Alzheimer's Disease (AD) pathology effectively with the only use of amyloid PET imaging modality from a brain-dedicated PET scanner. METHODS This study included 26 patients with amyloid PET images with 3 different radiotracers. All patients were acquired both on a PET/CT and a brain-dedicated PET scanner (CareMiBrain, CMB), from which 4 different reconstructions were implemented. A new pipeline was proposed and used for the PET image analysis based on the original Centiloid Scale processing pipeline, but with only PET images. The Youden's Index was employed to calculate the optimal cutoffs for diagnosis and evaluated by the AUC, accuracy, precision, and recall metrics. RESULTS The Centiloid Scale (CL) processing pipeline was validated with and without the use of MR images. The CL cutoffs for AD pathology diagnosis on the PET/CT and the 4 CMB reconstructions were 34.4 ± 2.2, 43.5 ± 3.5, 51.9 ± 12.5, 57.5 ± 6.8 and 41.8 ± 1.2 respectively. Overall, for these cutoffs all metrics obtained the maximum score. CONCLUSION The Centiloid scale applied to PET images allows for AD pathology diagnosis. The CMB scanner can be used with the Centiloid scale to automatically assist in the diagnosis of AD pathology, relieving the large burden of neurodegenerative diseases on a traditional PET/CT.
Collapse
Affiliation(s)
- Maria Teresa Gandia-Ferrero
- Biomedical Imaging Research Group (GIBI2(30)), La Fe Health Research Institute (IIS La Fe), Avenida Fernando Abril Martorell, València 46026, Spain.
| | - Irene Torres-Espallardo
- Biomedical Imaging Research Group (GIBI2(30)), La Fe Health Research Institute (IIS La Fe), Avenida Fernando Abril Martorell, València 46026, Spain; Nuclear Medicine Department, La Fe University and Polytechnic Hospital, Avenida Fernando Abril Martorell, València 46026, Spain
| | - Begoña Martínez-Sanchis
- Nuclear Medicine Department, La Fe University and Polytechnic Hospital, Avenida Fernando Abril Martorell, València 46026, Spain
| | - Enrique Muñoz
- Oncovision, Carrer de Jeroni de Montsoriu, 92, València 46022, Spain
| | | | - Pablo Sopena-Novales
- Nuclear Medicine Department, La Fe University and Polytechnic Hospital, Avenida Fernando Abril Martorell, València 46026, Spain
| | - Lourdes Álvarez-Sánchez
- Neurology Department, La Fe University and Polytechnic Hospital, Avenida Fernando Abril Martorell, València 46026, Spain
| | - Miquel Baquero-Toledo
- Neurology Department, La Fe University and Polytechnic Hospital, Avenida Fernando Abril Martorell, València 46026, Spain
| | - Luis Martí-Bonmatí
- Biomedical Imaging Research Group (GIBI2(30)), La Fe Health Research Institute (IIS La Fe), Avenida Fernando Abril Martorell, València 46026, Spain; Radiology Department, La Fe University and Polytechnic Hospital, Avenida Fernando Abril Martorell, València 46026, Spain
| |
Collapse
|
49
|
Katsumi Y, Howe IA, Eckbo R, Wong B, Quimby M, Hochberg D, McGinnis SM, Putcha D, Wolk DA, Touroutoglou A, Dickerson BC. Default mode network tau predicts future clinical decline in atypical early Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.17.24305620. [PMID: 38699357 PMCID: PMC11065041 DOI: 10.1101/2024.04.17.24305620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Identifying individuals with early stage Alzheimer's disease (AD) at greater risk of steeper clinical decline would allow professionals and loved ones to make better-informed medical, support, and life planning decisions. Despite accumulating evidence on the clinical prognostic value of tau PET in typical late-onset amnestic AD, its utility in predicting clinical decline in individuals with atypical forms of AD remains unclear. In this study, we examined the relationship between baseline tau PET signal and the rate of subsequent clinical decline in a sample of 48 A+/T+/N+ patients with mild cognitive impairment or mild dementia due to AD with atypical clinical phenotypes (Posterior Cortical Atrophy, logopenic variant Primary Progressive Aphasia, and amnestic syndrome with multi-domain impairment and age of onset < 65 years). All patients underwent structural magnetic resonance imaging (MRI), tau (18F-Flortaucipir) PET, and amyloid (either 18F-Florbetaben or 11C-Pittsburgh Compound B) PET scans at baseline. Each patient's longitudinal clinical decline was assessed by calculating the annualized change in the Clinical Dementia Rating Sum-of-Boxes (CDR-SB) scores from baseline to follow-up (mean time interval = 14.55 ± 3.97 months). Our sample of early atypical AD patients showed an increase in CDR-SB by 1.18 ± 1.25 points per year: t(47) = 6.56, p < .001, d = 0.95. These AD patients showed prominent baseline tau burden in posterior cortical regions including the major nodes of the default mode network, including the angular gyrus, posterior cingulate cortex/precuneus, and lateral temporal cortex. Greater baseline tau in the broader default mode network predicted faster clinical decline. Tau in the default mode network was the strongest predictor of clinical decline, outperforming baseline clinical impairment, tau in other functional networks, and the magnitude of cortical atrophy and amyloid burden in the default mode network. Overall, these findings point to the contribution of baseline tau burden within the default mode network of the cerebral cortex to predicting the magnitude of clinical decline in a sample of atypical early AD patients one year later. This simple measure based on a tau PET scan could aid the development of a personalized prognostic, monitoring, and treatment plan tailored to each individual patient, which would help clinicians not only predict the natural evolution of the disease but also estimate the effect of disease-modifying therapies on slowing subsequent clinical decline given the patient's tau burden while still early in the disease course.
Collapse
Affiliation(s)
- Yuta Katsumi
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Inola A Howe
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Ryan Eckbo
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Bonnie Wong
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Megan Quimby
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Daisy Hochberg
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Scott M McGinnis
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Center for Brain Mind Medicine, Department of Neurology, Brigham & Women's Hospital, Boston, MA 02115, USA
| | - Deepti Putcha
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Center for Brain Mind Medicine, Department of Neurology, Brigham & Women's Hospital, Boston, MA 02115, USA
| | - David A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexandra Touroutoglou
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Alzheimer's Disease Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Bradford C Dickerson
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Alzheimer's Disease Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| |
Collapse
|
50
|
Bischof GN, Jaeger E, Giehl K, Jessen F, Onur OA, O'Bryant S, Kara E, Weiss PH, Drzezga A. Cortical Tau Aggregation Patterns associated with Apraxia in Alzheimer's Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.09.24305535. [PMID: 38645131 PMCID: PMC11030486 DOI: 10.1101/2024.04.09.24305535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Objectives Apraxia is a core feature of Alzheimer's disease, but the pathomechanism of this characteristic symptom is not well understood. Here, we systematically investigated apraxia profiles in a well-defined group of patients with Alzheimer's disease (AD; N=32) who additionally underwent PET imaging with the second-generation tau PET tracer [18F]PI-2620. We hypothesized that specific patterns of tau pathology might be related to apraxic deficits. Methods Patients (N=32) with a biomarker-confirmed diagnosis of Alzheimer's disease were recruited in addition to a sample cognitively unimpaired controls (CU 1 ; N=41). Both groups underwent in-depth neuropsychological assessment of apraxia (Dementia Apraxia Screening Test; DATE and the Cologne Apraxia Screening; KAS). In addition, static PET imaging with [18F]PI-2620 was performed to assess tau pathology in the AD patients. To specifically investigate the association of apraxia with regional tau-pathology, we compared the PET-data from this group with an independent sample of amyloid-negative cognitively intact participants (CU 2; N=54) by generation of z-score-deviation maps as well as voxel- based multiple regression analyses. Results We identified significant clusters of tau-aggregation in praxis-related regions (e.g., supramarginal gyrus, angular gyrus, temporal, parietal and occipital regions) that were associated with apraxia. These regions were similar between the two apraxia assessments. No correlations between tau-tracer uptake in primary motor cortical or subcortical brain regions and apraxia were observed. Conclusions These results suggest that tau deposition in specific cortical brain regions may induce local neuronal dysfunction leading to a dose-dependent functional decline in praxis performance.
Collapse
|