1
|
Pan Y, Li L, Cao N, Liao J, Chen H, Zhang M. Advanced nano delivery system for stem cell therapy for Alzheimer's disease. Biomaterials 2025; 314:122852. [PMID: 39357149 DOI: 10.1016/j.biomaterials.2024.122852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/10/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
Alzheimer's Disease (AD) represents one of the most significant neurodegenerative challenges of our time, with its increasing prevalence and the lack of curative treatments underscoring an urgent need for innovative therapeutic strategies. Stem cells (SCs) therapy emerges as a promising frontier, offering potential mechanisms for neuroregeneration, neuroprotection, and disease modification in AD. This article provides a comprehensive overview of the current landscape and future directions of stem cell therapy in AD treatment, addressing key aspects such as stem cell migration, differentiation, paracrine effects, and mitochondrial translocation. Despite the promising therapeutic mechanisms of SCs, translating these findings into clinical applications faces substantial hurdles, including production scalability, quality control, ethical concerns, immunogenicity, and regulatory challenges. Furthermore, we delve into emerging trends in stem cell modification and application, highlighting the roles of genetic engineering, biomaterials, and advanced delivery systems. Potential solutions to overcome translational barriers are discussed, emphasizing the importance of interdisciplinary collaboration, regulatory harmonization, and adaptive clinical trial designs. The article concludes with reflections on the future of stem cell therapy in AD, balancing optimism with a pragmatic recognition of the challenges ahead. As we navigate these complexities, the ultimate goal remains to translate stem cell research into safe, effective, and accessible treatments for AD, heralding a new era in the fight against this devastating disease.
Collapse
Affiliation(s)
- Yilong Pan
- Department of Cardiology, Shengjing Hospital of China Medical University, Liaoning, 110004, China.
| | - Long Li
- Department of Neurosurgery, First Hospital of China Medical University, Liaoning, 110001, China.
| | - Ning Cao
- Army Medical University, Chongqing, 400000, China
| | - Jun Liao
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| | - Huiyue Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Liaoning, 110001, China.
| | - Meng Zhang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Liaoning, 110004, China.
| |
Collapse
|
2
|
Aumont E, Bedard MA, Bussy A, Arias JF, Tissot C, Hall BJ, Therriault J, Rahmouni N, Stevenson J, Servaes S, Macedo AC, Vitali P, Poltronetti NM, Fliaguine O, Trudel L, Gauthier S, Chakravarty MM, Rosa-Neto P. Hippocampal atrophy over two years in relation to tau, amyloid-β and memory in older adults. Neurobiol Aging 2025; 146:48-57. [PMID: 39631245 DOI: 10.1016/j.neurobiolaging.2024.11.007] [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: 05/31/2024] [Revised: 09/27/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024]
Abstract
In this longitudinal brain imaging study, we aimed to characterize hippocampal tau accumulation and subfield atrophy relative to cortical amyloid-β and memory performance. We measured tau-PET in regions associated with Braak stages I to VI, global amyloid-PET burden, hippocampal subfield volumes and memory assessments from 173 participants aged 55-85. Eighty-six of these participants were tested again two years later. Tau-PET change in the Braak II region, corresponding to the hippocampus and the entorhinal cortex, was significantly associated with the cornu ammonis 1 (CA1) atrophy and memory score. This CA1 atrophy did not significantly mediate the association between tau and memory, nor did global amyloid-PET burden correlate with tau-PET changes in the Braak II region. Longitudinal hippocampal tau accumulation is amyloid-β-independent and co-localized with subfield atrophy. As tau-associated memory decline seems to be independent from hippocampal atrophy, other mechanisms could contribute to the deficit.
Collapse
Affiliation(s)
- Etienne Aumont
- NeuroQAM Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC H2X 3P2, Canada; McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| | - Marc-André Bedard
- NeuroQAM Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC H2X 3P2, Canada; McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| | - Aurélie Bussy
- Cerebral Imaging Center, Douglas Research Center, Montreal, QC H4H 1R3, Canada; Computational Brain Anatomy (CoBrALab) Laboratory, Montreal, QC H4H 1R3, Canada
| | - Jaime Fernandez Arias
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | - Cecile Tissot
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada; Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
| | - Brandon J Hall
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | - Joseph Therriault
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | - Nesrine Rahmouni
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | - Jenna Stevenson
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | - Stijn Servaes
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | - Arthur C Macedo
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | - Paolo Vitali
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | | | - Olga Fliaguine
- NeuroQAM Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC H2X 3P2, Canada
| | - Lydia Trudel
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | - Serge Gauthier
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | - Mallar M Chakravarty
- Cerebral Imaging Center, Douglas Research Center, Montreal, QC H4H 1R3, Canada; Computational Brain Anatomy (CoBrALab) Laboratory, Montreal, QC H4H 1R3, Canada; Department of Psychiatry, McGill University, Montreal, QC H3A 1A1, Canada
| | - Pedro Rosa-Neto
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada.
| |
Collapse
|
3
|
Chu H, Huang C, Xie F, Guo Q. The Association Between Constipation and Positron Emission Tomography and Blood-Based Biomarkers in Older Cognitively Unimpaired Adults with Higher Amyloid-β Burden. Neurol Ther 2024; 13:1701-1715. [PMID: 39436582 PMCID: PMC11541973 DOI: 10.1007/s40120-024-00666-x] [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: 08/21/2024] [Accepted: 09/19/2024] [Indexed: 10/23/2024] Open
Abstract
INTRODUCTION Constipation may be linked to cognitive decline and a higher risk of Alzheimer's disease (AD). We aimed to investigate the association between constipation and positron emission tomography (PET) and blood-based AD biomarkers in older cognitively unimpaired (CU) adults with higher Aβ burden. METHODS Constipation was diagnosed according to Rome IV criteria and the severity of constipation was evaluated by using a validated self-reported questionnaire. The participants underwent the examination of plasma AD biomarkers and 18F-florbetapir PET and 18F-MK6240 PET scans; the latter was only performed in the validation cohort. Correlation and multiple linear regression analyses were used to investigate the association between constipation and AD biomarkers. RESULTS Two cohorts were included in our study. A total of 404 older participants with 126 of whom Aβ-PET positive were enrolled in the development cohort. Multiple linear regression analysis showed constipation was associated with plasma t-Tau, p-Tau-181, and neurofilament light chain (NfL) in participants with Aβ-PET (+). Meanwhile, no/mild constipation was associated with lower Aβ-PET standard uptake value ratio. The association between constipation and plasma biomarkers was different in the subgroups stratified by age, sex and APOE ε4 genotype. The above associations were further validated in the validation cohort containing 36 Aβ-PET (+) participants. Importantly, no/mild constipation was associated with less Tau burden evaluated by 18F-MK6240 PET Braak stages. CONCLUSION Our data indicate that no/mild constipation may be associated with lower plasma t-Tau, p-Tau-181, and NfL as well as less Aβ and Tau burden in older CU adults with Aβ deposition. Improving constipation and being away from defecation disorders may help reduce the risk of AD development.
Collapse
Affiliation(s)
- Heling Chu
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600, Yi Shan Road, Shanghai, China
| | - Chuyi Huang
- Health Management Center, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, No. 12 Mid Wulumuqi Road, Shanghai, China.
| | - Qihao Guo
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600, Yi Shan Road, Shanghai, China.
| |
Collapse
|
4
|
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
|
5
|
Cui L, Zhang Z, Tu YY, Wang M, Guan YH, Li YH, Xie F, Guo QH. Association of precuneus Aβ burden with default mode network function. Alzheimers Dement 2024. [PMID: 39559982 DOI: 10.1002/alz.14380] [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: 08/05/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 11/20/2024]
Abstract
INTRODUCTION It remains unclear whether the local amyloid-beta (Aβ) burden in key regions within the default mode network (DMN) affects network and cognitive functions. METHODS Participants included 1002 individuals from the Chinese Preclinical Alzheimer's Disease Study cohort who underwent 18F-florbetapir positron emission tomography resting-state functional magnetic resonance imaging scanning and neuropsychological tests. The correlations between precuneus (PRC) Aβ burden, DMN function, and cognitive function were investigated. RESULTS In individuals with high PRC Aβ burden, there is a bidirectional relationship between DMN local function or functional connectivity and PRC Aβ deposition across various cognitive states, which is also linked to cognitive function. Even below the PRC Aβ threshold, DMN function remains related to PRC Aβ deposition and cognitive performance. DISCUSSION The findings reveal the critical role of PRC Aβ deposition in disrupting neural networks associated with cognitive decline and the necessity of early detection and monitoring of PRC Aβ deposition. HIGHLIGHTS Precuneus (PRC) Aβ burden impacts DMN function in different cognitive stages. High PRC Aβ burden is linked to early neural compensation and subsequent dysfunction. Low PRC Aβ burden correlates with neural changes before significant Aβ accumulation. Changes in DMN function and connectivity provide insights into AD progression. Early detection of regional Aβ burden can help monitor the risk of cognitive decline.
Collapse
Affiliation(s)
- Liang Cui
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Zhang
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - You-Yi Tu
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Wang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Yi-Hui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yue-Hua Li
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qi-Hao Guo
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Rea Reyes RE, Cody KA, Wilson RE, Zetterberg H, Chin NA, Jonaitis EM, Bahr M, Mandel O, Wintlend M, Bendlin BB, Okonkwo OC, Clark LR, Zammit M, Asthana S, Christian BT, Betthauser TJ, Eisenmenger L, Langhough RE, Johnson SC. Visual read of [F-18]florquinitau PET that includes and extends beyond the mesial temporal lobe is associated with increased plasma pTau217 and cognitive decline in a cohort that is enriched with risk for Alzheimer's disease. Alzheimers Dement 2024. [PMID: 39560002 DOI: 10.1002/alz.14406] [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: 08/08/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/20/2024]
Abstract
INTRODUCTION Patterns of signal from tau positron emission tomography (tau-PET) confined to the medial temporal lobe (MTL) or extended into the neocortex may be relevant for Alzheimer's disease (AD) research if they are linked to differential biomarker levels and cognitive decline. METHODS Visual assessment of Tau-PET [F-18]florquinitau (FQT) exams from 728 initially non-demented older adults yielded four uptake groups: tau-negative (T-), MTL-only (T+MTL), neocortex-only (T+Neo), or both (T+MTL&Neo). Mixed effects models assessed group differences in retrospective cognitive and plasma pTau217 trajectories. RESULTS T+MTL&Neo was the most common T+ group (n = 97; 93% A+) and exhibited the sharpest worsening in cognitive and pTau217 trajectories before tau PET. DISCUSSION The T+MTL&Neo category represents an intermediate to advanced stage of AD preceded by rising ptau217 and progressive cognitive decline. The pTau217 finding suggests that A+, T+ in MTL or neocortex could represent early AD stages, with a higher likelihood of progressing to more advanced stages. HIGHLIGHTS Visual assessments of Tau-PET FQT revealed four distinct uptake groups: tau-negative (T-), MTL-only (T+MTL), neocortex-only (T+Neo), or both (T+MTL&Neo). Amyloid positive participants in the T+MTL and T+MTL&Neo categories showed a retrospectively faster decline in their cognitive trajectories, and a sharper increase in pTau217 levels in plasma, compared to T-. The T+MTL&Neo group displayed sharper trajectories compared with the other Tau positive groups in both their cognitive scores and pTau217 plasma levels. Our results suggest that participants with Tau present in both MTL and neocortex represent an intermediate to advanced stage of AD, whereas participants with signals confined to either MTL or neocortex could represent earlier AD stages.
Collapse
Affiliation(s)
- Ramiro Eduardo Rea Reyes
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Karly A Cody
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, California, USA
| | - Rachael E Wilson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Henrik Zetterberg
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Science Park, Hong Kong, China
| | - Nathaniel A Chin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Erin M Jonaitis
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Melissa Bahr
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Olivia Mandel
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Madilynn Wintlend
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Ozioma C Okonkwo
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Lindsay R Clark
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Matt Zammit
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Bradley T Christian
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Tobey J Betthauser
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Laura Eisenmenger
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Rebecca E Langhough
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
7
|
Woo MS, Therriault J, Jonaitis EM, Wilson R, Langhough RE, Rahmouni N, Benedet AL, Ashton NJ, Tissot C, Lantero-Rodriguez J, Macedo AC, Servaes S, Wang YT, Arias JF, Hosseini SA, Betthauser TJ, Lussier FZ, Hopewell R, Triana-Baltzer G, Kolb HC, Jeromin A, Kobayashi E, Massarweh G, Friese MA, Klostranec J, Vilali P, Pascoal TA, Gauthier S, Zetterberg H, Blennow K, Johnson SC, Rosa-Neto P. Identification of late-stage tau accumulation using plasma phospho-tau217. EBioMedicine 2024; 109:105413. [PMID: 39500009 PMCID: PMC11570195 DOI: 10.1016/j.ebiom.2024.105413] [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: 04/10/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Blood-based disease staging across the Alzheimer's disease (AD) continuum holds the promise to identify individuals that profit from disease-modifying therapies. We set out to identify Braak V+ (Braak V and/or VI) tau PET-positive individuals within amyloid-β (Aβ)-positive individuals using plasma biomarkers. METHODS In this cross-sectional study, we assessed 289 individuals from the TRIAD cohort and 306 individuals from the WRAP study across the AD continuum. The participants were evaluated by amyloid-PET with [18F]AZD4694 or [11C]PiB and tau-PET with [18F]MK6240 and measured plasma levels included total tau, phospho-tau isoforms (pTau) pTau-181, pTau-217, pTau-231, and N-terminal tau (NTA-tau). We evaluated the performances of plasma biomarkers using different analytic platforms to predict Braak V+ positivity in Aβ+ individuals. FINDINGS Highest associations with Braak V+ tau positivity in Aβ+ individuals were found for plasma pTau-217+Janssen (AUC [CI95%] = 0.97 [0.94, 1.0]) and ALZpath pTau-217 (AUC [CI95%] = 0.93 [0.86, 1.0]) in TRIAD. Plasma ALZpath pTau-217 separated Braak V+ tau PET-positive individuals in the WRAP longitudinal study (AUC [CI95%] = 0.97 [0.94, 1.0]). INTERPRETATION Thus, we demonstrate that using adjusted cut-offs, plasma pTau-217 identifies individuals with later Braak stage tau accumulation which will be helpful to stratify patients for treatments and clinical studies. FUNDING This research is supported by the Weston Brain Institute, Canadian Institutes of Health Research (CIHR) [MOP-11-51-31; RFN 152985, 159815, 162303], Canadian Consortium of Neurodegeneration and Aging (CCNA; MOP-11-51-31 -team 1), the Alzheimer's Association [NIRG-12-92090, NIRP-12-259245], Brain Canada Foundation (CFI Project 34874; 33397), the Fonds de Recherche du Québec-Santé (FRQS; Chercheur Boursier, 2020-VICO-279314). P.R-N and SG are members of the CIHR-CCNA Canadian Consortium of Neurodegeneration in Aging. Colin J. Adair Charitable Foundation.
Collapse
Affiliation(s)
- Marcel S Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Centre Hamburg Eppendorf, 20251, Hamburg, Germany; Department of Neurology, University Medical Centre Hamburg Eppendorf, 20251, Hamburg, Germany; Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, QC H4H 1R3, Canada.
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, QC H3A 1A1, Canada
| | - Erin M Jonaitis
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53726, USA
| | - Rachael Wilson
- Wisconsin Alzheimer's Disease Research Centre, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53726, USA
| | - Rebecca E Langhough
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53726, USA
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, QC H4H 1R3, Canada
| | - Andrea Lessa Benedet
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, S-431 80, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, S-431 80, Mölndal, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Cécile Tissot
- Molecular Biophysics and Integrated Bioimaging Department, Lawrence Berkeley National Laboratory, Berkeley, 510, CA, USA
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, S-431 80, Mölndal, Sweden
| | - Arthur C Macedo
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, QC H3A 1A1, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, QC H4H 1R3, Canada
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, QC H3A 1A1, Canada
| | - Jaime Fernandez Arias
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, QC H4H 1R3, Canada
| | - Seyyed Ali Hosseini
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, QC H4H 1R3, Canada
| | - Tobey J Betthauser
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53726, USA
| | - Firoza Z Lussier
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Robert Hopewell
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, QC H3A 1A1, Canada
| | - Gallen Triana-Baltzer
- Neuroscience Biomarkers, Johnson and Johnson Medical Innovation (formerly Janssen Research & Development), La Jolla, CA, 92121, USA
| | - Hartmuth C Kolb
- Neuroscience Biomarkers, Johnson and Johnson Medical Innovation (formerly Janssen Research & Development), La Jolla, CA, 92121, USA
| | | | - Eliane Kobayashi
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, S-431 80, Mölndal, Sweden
| | - Gassan Massarweh
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, QC H3A 1A1, Canada
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Centre Hamburg Eppendorf, 20251, Hamburg, Germany
| | - Jesse Klostranec
- Montreal Neurologic Institute and Hospital, Department of Diagnostic and Interventional Neuroradiology, McGill University Health Centre, 3801 Rue University, Montréal, QC, H3A 2B4, Canada
| | - Paolo Vilali
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, QC H3A 1A1, Canada
| | - Tharick A Pascoal
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, QC H3A 1A1, Canada
| | - Henrik Zetterberg
- Wisconsin Alzheimer's Disease Research Centre, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53726, USA; Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, S-431 80, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1E 6BT, UK; UK Dementia Research Institute at UCL, London, WC1E 6BT, UK; Hong Kong Centre for Neurodegenerative Diseases, Clear Water Bay, 518172 Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, S-431 80, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80, Mölndal, Sweden
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53726, USA; Wisconsin Alzheimer's Disease Research Centre, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53726, USA
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, QC H3A 1A1, Canada.
| |
Collapse
|
8
|
Vermeiren MR, Somsen J, Luurtsema G, Reesink FE, Verwey NA, Hempenius L, Tolboom N, Biessels GJ, Biesbroek JM, Vernooij MW, Veldhuijzen van Zanten SEM, Seelaar H, Coomans EM, Teunissen CE, Lemstra AW, van Harten AC, Visser LNC, van der Flier WM, van de Giessen E, Ossenkoppele R. The impact of tau-PET in a selected memory clinic cohort: rationale and design of the TAP-TAU study. Alzheimers Res Ther 2024; 16:230. [PMID: 39427210 PMCID: PMC11490118 DOI: 10.1186/s13195-024-01588-4] [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] [Accepted: 09/29/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Tau-PET is a diagnostic tool with high sensitivity and specificity for discriminating Alzheimer's disease (AD) dementia from other neurodegenerative disorders in well-controlled research environments. The role of tau-PET in real-world clinical practice, however, remains to be established. The aim of the TAP-TAU study is therefore to investigate the impact of tau-PET in clinical practice. METHODS TAP-TAU is a prospective, longitudinal multi-center study in 300 patients (≥ 50 years old) with mild cognitive impairment or mild dementia across five Dutch memory clinics. Patients are eligible if diagnostic certainty is < 85% after routine dementia screening and if the differential diagnosis includes AD. More specifically, we will include patients who (i) are suspected of having mixed pathology (e.g., AD and vascular pathology), (ii) have an atypical clinical presentation, and/or (iii) show conflicting or inconclusive outcomes on other tests (e.g., magnetic resonance imaging or cerebrospinal fluid). Participants will undergo a [18F]flortaucipir tau-PET scan, blood-based biomarker sampling, and fill out questionnaires on patient reported outcomes and experiences. The primary outcomes are change (pre- versus post- tau-PET) in diagnosis, diagnostic certainty, patient management and patient anxiety and uncertainty. Secondary outcome measures are head-to-head comparisons between tau-PET and less invasive and lower cost diagnostic tools such as novel blood-based biomarkers and artificial intelligence-based classifiers. RESULTS TAP-TAU has been approved by the Medical Ethics Committee of the Amsterdam UMC. The first participant is expected to be included in October 2024. CONCLUSIONS In TAP-TAU, we will investigate the added clinical value of tau-PET in a real-world clinical setting, including memory clinic patients with diagnostic uncertainty after routine work-up. Findings of our study may contribute to recommendations regarding which patients would benefit most from assessment with tau-PET. This study is timely in the dawning era of disease modifying treatments as an accurate etiological diagnosis becomes increasingly important. TRIAL REGISTRATION This trial is registered and authorized on December 21st, 2023 in EU Clinical Trials with registration number 2023-505430-10-00.
Collapse
Affiliation(s)
- Marie R Vermeiren
- Alzheimer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands.
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands.
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands.
| | - Joost Somsen
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Gert Luurtsema
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Fransje E Reesink
- Department of Neurology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Nicolaas A Verwey
- Department of Neurology, Medical Center Leeuwarden, Leeuwarden, Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands
| | | | - Nelleke Tolboom
- Department of Radiology and Nuclear Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Geert Jan Biessels
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - J Matthijs Biesbroek
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Neurology, Diakonessenhuis Hospital, Utrecht, Netherlands
| | - Meike W Vernooij
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | - Harro Seelaar
- Department of Neurology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Emma M Coomans
- Alzheimer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands
| | | | - Afina W Lemstra
- Alzheimer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
| | - Argonde C van Harten
- Alzheimer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
| | - Leonie N C Visser
- Alzheimer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Psychology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands
- Epidemiology and Data Science, Amsterdam UMC, Amsterdam, Netherlands
| | - Elsmarieke van de Giessen
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands.
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands.
- Clinical Memory Research Unit, Lund University, Lund, Sweden.
| |
Collapse
|
9
|
Zhang S, Crossley CA, Yuan Q. Neuronal Vulnerability of the Entorhinal Cortex to Tau Pathology in Alzheimer's Disease. Br J Biomed Sci 2024; 81:13169. [PMID: 39435008 PMCID: PMC11491395 DOI: 10.3389/bjbs.2024.13169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024]
Abstract
This review delves into the entorhinal cortex (EC) as a central player in the pathogenesis of Alzheimer's Disease (AD), emphasizing its role in the accumulation and propagation of tau pathology. It elucidates the multifaceted functions of the EC, encompassing memory formation, spatial navigation, and olfactory processing, while exploring how disruptions in these processes contribute to cognitive decline in AD. The review discusses the intricate interplay between tau pathology and EC vulnerability, highlighting how alterations in neuronal firing patterns and synaptic function within the EC exacerbate cognitive impairments. Furthermore, it elucidates how specific neuronal subtypes within the EC exhibit differential susceptibility to tau-induced damage, contributing to disease progression. Early detection methods, such as imaging techniques and assessments of EC blood flow, are examined as potential tools for identifying tau pathology in the preclinical stages of AD. These approaches offer promise for improving diagnostic accuracy and enabling timely intervention. Therapeutic strategies targeting tau pathology within the EC are explored, including the clearance of pathological tau aggregates and the inhibition of tau aggregation processes. By understanding the molecular and cellular mechanisms underlying EC vulnerability, researchers can develop more targeted and effective interventions to slow disease progression. The review underscores the importance of reliable biomarkers to assess disease progression and therapeutic efficacy in clinical trials targeting the EC. Ultimately, it aims to contribute to the development of more effective management strategies for AD, emphasizing the translation of research findings into clinical practice to address the growing societal burden of the disease.
Collapse
Affiliation(s)
| | - Chelsea Ann Crossley
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| | | |
Collapse
|
10
|
Vermeiren MR, Calandri IL, van der Flier WM, van de Giessen E, Ossenkoppele R. Survey among experts on the future role of tau-PET in clinical practice and trials. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e70033. [PMID: 39583643 PMCID: PMC11582687 DOI: 10.1002/dad2.70033] [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: 07/03/2024] [Accepted: 10/02/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Recent advancements in Alzheimer's disease (AD) biomarker research and clinical trials prompt reflection on the value and consequently appropriate use of tau positron emission tomography (tau-PET) in the future. METHODS We conducted an online survey among dementia and PET experts worldwide to investigate the anticipated future role of tau-PET in clinical practice and trials. RESULTS Two hundred sixty-eight dementia experts, comprising 143 clinicians and 121 researchers, covering six continents participated. The vast majority (90%) fostered a positive attitude toward the added value of tau-PET in clinical practice, particularly for staging, diagnosing, monitoring, and prognostication in a cognitively impaired memory clinic population. Experts anticipated an important role for tau-PET for participant selection (76%-100%) and measuring endpoints (75%-97%), in both anti-amyloid and anti-tau drug trials. DISCUSSION Our global survey study shows that dementia experts envision an important role for tau-PET in the future, both in clinical practice and in drug trials, beyond current guidelines and practices. Highlights Dementia experts envision an important role for tau-PET in the future.Experts indicate that a tau-PET scan could influence patient management.Experts anticipate the utility of tau-PET for participant selection and endpoints in drug trials.There is a gap between the anticipated usefulness of tau-PET and current clinical practices.
Collapse
Affiliation(s)
- Marie R. Vermeiren
- Alzheimer Center Amsterdam, NeurologyVrije Universiteit AmsterdamAmsterdam UMCAmsterdamThe Netherlands
- Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMCAmsterdamThe Netherlands
- Amsterdam NeuroscienceBrain ImagingAmsterdamThe Netherlands
| | - Ismael L. Calandri
- Alzheimer Center Amsterdam, NeurologyVrije Universiteit AmsterdamAmsterdam UMCAmsterdamThe Netherlands
- Department of Cognitive NeurologyFleniBuenos AiresArgentina
- Amsterdam NeuroscienceNeurodegenerationAmsterdamThe Netherlands
| | - Wiesje M. van der Flier
- Alzheimer Center Amsterdam, NeurologyVrije Universiteit AmsterdamAmsterdam UMCAmsterdamThe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamThe Netherlands
- Epidemiology and Data ScienceVrije Universiteit Amsterdam, Amsterdam UMCAmsterdamThe Netherlands
| | - Elsmarieke van de Giessen
- Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMCAmsterdamThe Netherlands
- Amsterdam NeuroscienceBrain ImagingAmsterdamThe Netherlands
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, NeurologyVrije Universiteit AmsterdamAmsterdam UMCAmsterdamThe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamThe Netherlands
- Clinical Memory Research UnitLund UniversityLundSweden
| |
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
|
Pascoal TA, Aguzzoli CS, Lussier FZ, Crivelli L, Suemoto CK, Fortea J, Rosa-Neto P, Zimmer ER, Ferreira PCL, Bellaver B. Insights into the use of biomarkers in clinical trials in Alzheimer's disease. EBioMedicine 2024; 108:105322. [PMID: 39366844 DOI: 10.1016/j.ebiom.2024.105322] [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: 02/26/2024] [Revised: 08/12/2024] [Accepted: 08/21/2024] [Indexed: 10/06/2024] Open
Abstract
Biomarkers have been instrumental in population selection and disease monitoring in clinical trials of recently FDA-approved drugs targeting amyloid-β to slow the progression of Alzheimer's disease (AD). As new therapeutic strategies and biomarker techniques emerge, the importance of biomarkers in drug development is growing exponentially. In this emerging landscape, biomarkers are expected to serve a wide range of contexts of use in clinical trials focusing on AD and related dementias. The joint FDA-NIH BEST (Biomarkers, EndpointS, and other Tools) framework provides standardised terminology to facilitate communication among stakeholders in this increasingly complex field. This review explores various applications of biomarkers relevant to AD clinical trials, using the BEST resource as a reference. For simplicity, we predominantly provide contextual characterizations of biomarkers use from the perspective of drugs targeting amyloid-β and tau proteins. However, general definitions and concepts can be extrapolated to other targets.
Collapse
Affiliation(s)
- Tharick A Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | | | - Firoza Z Lussier
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Lucía Crivelli
- Department of Cognitive Neurology, Fleni, Buenos Aires, Argentina
| | - Claudia K Suemoto
- Division of Geriatrics, University of São Paulo Medical School, São Paulo, Brazil
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
| | - Eduardo R Zimmer
- Brain Institute of Rio Grande do Sul, PUCRS, Porto Alegre, Brazil; Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil; Graduate Program in Biological Sciences, Biochemistry (PPGBioq), and Pharmacology and Therapeutics (PPGFT), Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Pamela C L Ferreira
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Bruna Bellaver
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
13
|
Karlsson L, Vogel J, Arvidsson I, Åström K, Strandberg O, Seidlitz J, Bethlehem RAI, Stomrud E, Ossenkoppele R, Ashton NJ, Zetterberg H, Blennow K, Palmqvist S, Smith R, Janelidze S, Joie RL, Rabinovici GD, Binette AP, Mattsson-Carlgren N, Hansson O. A machine learning-based prediction of tau load and distribution in Alzheimer's disease using plasma, MRI and clinical variables. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.31.24308264. [PMID: 38853877 PMCID: PMC11160861 DOI: 10.1101/2024.05.31.24308264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Tau positron emission tomography (PET) is a reliable neuroimaging technique for assessing regional load of tau pathology in the brain, commonly used in Alzheimer's disease (AD) research and clinical trials. However, its routine clinical use is limited by cost and accessibility barriers. Here we explore using machine learning (ML) models to predict clinically useful tau-PET composites from low-cost and non-invasive features, e.g., basic clinical variables, plasma biomarkers, and structural magnetic resonance imaging (MRI). Results demonstrated that models including plasma biomarkers yielded the most accurate predictions of tau-PET burden (best model: R-squared=0.66-0.68), with especially high contribution from plasma P-tau217. In contrast, MRI variables stood out as best predictors (best model: R-squared=0.28-0.42) of asymmetric tau load between the two hemispheres (an example of clinically relevant spatial information). The models showed high generalizability to external test cohorts with data collected at multiple sites. Based on these results, we also propose a proof-of-concept two-step classification workflow, demonstrating how the ML models can be translated to a clinical setting. This study uncovers current potential in predicting tau-PET information from scalable cost-effective variables, which could improve diagnosis and prognosis of AD.
Collapse
Affiliation(s)
- Linda Karlsson
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Jacob Vogel
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Department of Clinical Sciences, SciLifeLab, Lund University, Lund, Sweden
| | - Ida Arvidsson
- Centre for Mathematical Sciences, Lund University, Lund, Sweden
| | - Kalle Åström
- Centre for Mathematical Sciences, Lund University, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Jakob Seidlitz
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, 19104 USA
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104 USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Richard A. I. Bethlehem
- University of Cambridge, Department of Psychology, Cambridge Biomedical Campus, Cambridge, CB2 3EB, UK
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, Netherlands
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience, King’s College London, London, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Renaud La Joie
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Gil D. Rabinovici
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
14
|
Kang DW, Wang SM, Um YH, Kim S, Kim T, Kim D, Lee CU, Lim HK. Transcranial direct current stimulation and neuronal functional connectivity in MCI: role of individual factors associated to AD. Front Psychiatry 2024; 15:1428535. [PMID: 39224475 PMCID: PMC11366601 DOI: 10.3389/fpsyt.2024.1428535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/11/2024] [Indexed: 09/04/2024] Open
Abstract
Background Alzheimer's disease (AD) encompasses a spectrum that may progress from mild cognitive impairment (MCI) to full dementia, characterized by amyloid-beta and tau accumulation. Transcranial direct current stimulation (tDCS) is being investigated as a therapeutic option, but its efficacy in relation to individual genetic and biological risk factors remains underexplored. Objective To evaluate the effects of a two-week anodal tDCS regimen on the left dorsolateral prefrontal cortex, focusing on functional connectivity changes in neural networks in MCI patients resulting from various possible underlying disorders, considering individual factors associated to AD such as amyloid-beta deposition, APOE ϵ4 allele, BDNF Val66Met polymorphism, and sex. Methods In a single-arm prospective study, 63 patients with MCI, including both amyloid-PET positive and negative cases, received 10 sessions of tDCS. We assessed intra- and inter-network functional connectivity (FC) using fMRI and analyzed interactions between tDCS effects and individual factors associated to AD. Results tDCS significantly enhanced intra-network FC within the Salience Network (SN) and inter-network FC between the Central Executive Network and SN, predominantly in APOE ϵ4 carriers. We also observed significant sex*tDCS interactions that benefited inter-network FC among females. Furthermore, the effects of multiple modifiers, particularly the interaction of the BDNF Val66Met polymorphism and sex, were evident, as demonstrated by increased intra-network FC of the SN in female Met non-carriers. Lastly, the effects of tDCS on FC did not differ between the group of 26 MCI patients with cerebral amyloid-beta deposition detected by flutemetamol PET and the group of 37 MCI patients without cerebral amyloid-beta deposition. Conclusions The study highlights the importance of precision medicine in tDCS applications for MCI, suggesting that individual genetic and biological profiles significantly influence therapeutic outcomes. Tailoring interventions based on these profiles may optimize treatment efficacy in early stages of AD.
Collapse
Affiliation(s)
- Dong Woo Kang
- Department of Psychiatry, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sheng-Min Wang
- Department of Psychiatry, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoo Hyun Um
- Department of Psychiatry, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sunghwan Kim
- Department of Psychiatry, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - TaeYeong Kim
- Research Institute, NEUROPHET Inc., Seoul, Republic of Korea
| | - Donghyeon Kim
- Research Institute, NEUROPHET Inc., Seoul, Republic of Korea
| | - Chang Uk Lee
- Department of Psychiatry, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyun Kook Lim
- Department of Psychiatry, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Research Institute, NEUROPHET Inc., Seoul, Republic of Korea
| |
Collapse
|
15
|
Chu H, Huang C, Guan Y, Xie F, Chen M, Guo Q. The associations between nutritional status and physical frailty and Alzheimer's disease plasma biomarkers in older cognitively unimpaired adults with positive of amyloid-β PET. Clin Nutr 2024; 43:1647-1656. [PMID: 38810424 DOI: 10.1016/j.clnu.2024.05.024] [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: 02/01/2024] [Revised: 05/06/2024] [Accepted: 05/12/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND & AIMS It has been revealed good nutritional status and no physical frailty, which are modifiable lifestyle factors, are linked to less cognitive decline and a lower risk of Alzheimer's disease (AD). We aimed to investigate the associations between nutritional status and physical frailty and plasma AD biomarkers, especially the Tau-associated biomarkers in older cognitively unimpaired (CU) adults with higher β-amyloid (Aβ) burden. METHODS The nutritional status and physical frailty were assessed via Mini-Nutritional Assessment Short-Form (MNA-SF) and Fried frailty index. The participants underwent the examination of plasma AD biomarkers and 18F-florbetapir PET scan as well as 18F-MK6240 PET in the validation cohort. Correlation and multiple linear regression analyses were used to investigate the associations between nutritional status and frailty and AD biomarkers. RESULTS Two cohorts were included in our study. A total of 129 participants with Aβ-PET positive were enrolled in the development cohort. Multiple linear regression analysis showed MNA-SF scores, normal nutritional status, Fried frailty index scores, frailty and some domains of frailty including weight loss, maximal grip strength and exhaustion were associated with plasma p-Tau-181. Furthermore, weight loss, Fried frailty index scores and frailty were associated with higher Aβ-PET standard uptake value ratio. We further performed subgroup analyses stratified by age, sex and apolipoprotein E ε4 genotype to investigate the beneficial characteristics of nutrition and frailty in the special subgroups. Validation cohort contained 38 Aβ-PET positive participants. MNA-SF scores, normal nutritional status, Fried frailty index scores and frailty were associated with Tau burden evaluated by 18F-MK6240 PET Braak-like stages. CONCLUSIONS Our data indicates that normal nutritional status and no physical frailty may be associated with expected trend of plasma AD biomarkers, especially less Tau pathology in older CU adults with Aβ deposition. Adjusting to these characteristics of nutrition and physical frailty may help reduce the risk of AD development.
Collapse
Affiliation(s)
- Heling Chu
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuyi Huang
- Health Management Center, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Meirong Chen
- Department of Neurorehabilitation High Dependency Unit, Jiangwan Hospital, Shanghai, China.
| | - Qihao Guo
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
16
|
Sanchez-Rodriguez LM, Khan AF, Adewale Q, Bezgin G, Therriault J, Fernandez-Arias J, Servaes S, Rahmouni N, Tissot C, Stevenson J, Jiang H, Chai X, Carbonell F, Rosa-Neto P, Iturria-Medina Y. In-vivo neuronal dysfunction by Aβ and tau overlaps with brain-wide inflammatory mechanisms in Alzheimer's disease. Front Aging Neurosci 2024; 16:1383163. [PMID: 38966801 PMCID: PMC11223503 DOI: 10.3389/fnagi.2024.1383163] [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: 02/07/2024] [Accepted: 05/09/2024] [Indexed: 07/06/2024] Open
Abstract
The molecular mechanisms underlying neuronal dysfunction in Alzheimer's disease (AD) remain uncharacterized. Here, we identify genes, molecular pathways and cellular components associated with whole-brain dysregulation caused by amyloid-beta (Aβ) and tau deposits in the living human brain. We obtained in-vivo resting-state functional MRI (rs-fMRI), Aβ- and tau-PET for 47 cognitively unimpaired and 16 AD participants from the Translational Biomarkers in Aging and Dementia cohort. Adverse neuronal activity impacts by Aβ and tau were quantified with personalized dynamical models by fitting pathology-mediated computational signals to the participant's real rs-fMRIs. Then, we detected robust brain-wide associations between the spatial profiles of Aβ-tau impacts and gene expression in the neurotypical transcriptome (Allen Human Brain Atlas). Within the obtained distinctive signature of in-vivo neuronal dysfunction, several genes have prominent roles in microglial activation and in interactions with Aβ and tau. Moreover, cellular vulnerability estimations revealed strong association of microglial expression patterns with Aβ and tau's synergistic impact on neuronal activity (q < 0.001). These results further support the central role of the immune system and neuroinflammatory pathways in AD pathogenesis. Neuronal dysregulation by AD pathologies also associated with neurotypical synaptic and developmental processes. In addition, we identified drug candidates from the vast LINCS library to halt or reduce the observed Aβ-tau effects on neuronal activity. Top-ranked pharmacological interventions target inflammatory, cancer and cardiovascular pathways, including specific medications undergoing clinical evaluation in AD. Our findings, based on the examination of molecular-pathological-functional interactions in humans, may accelerate the process of bringing effective therapies into clinical practice.
Collapse
Affiliation(s)
- Lazaro M. Sanchez-Rodriguez
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, Montreal, QC, Canada
| | - Ahmed F. Khan
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, Montreal, QC, Canada
| | - Quadri Adewale
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, Montreal, QC, Canada
| | - Gleb Bezgin
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Joseph Therriault
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Jaime Fernandez-Arias
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Stijn Servaes
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Nesrine Rahmouni
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Cécile Tissot
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
- Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Jenna Stevenson
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Hongxiu Jiang
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
| | - Xiaoqian Chai
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
| | | | - Pedro Rosa-Neto
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Yasser Iturria-Medina
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, Montreal, QC, Canada
| |
Collapse
|
17
|
Ossenkoppele R, Salvadó G, Janelidze S, Binette AP, Bali D, Karlsson L, Palmqvist S, Mattsson-Carlgren N, Stomrud E, Therriault J, Rahmouni N, Rosa-Neto P, Coomans EM, van de Giessen E, van der Flier WM, Teunissen CE, Jonaitis EM, Johnson SC, Villeneuve S, Benzinger TL, Schindler SE, Bateman RJ, Doecke JD, Doré V, Feizpour A, Masters CL, Rowe C, Wiste HJ, Petersen RC, Jack CR, Hansson O. Prediction of future cognitive decline among cognitively unimpaired individuals using measures of soluble phosphorylated tau or tau tangle pathology. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.12.24308824. [PMID: 38947004 PMCID: PMC11213114 DOI: 10.1101/2024.06.12.24308824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Plasma p-tau217 and Tau-PET are strong prognostic biomarkers in Alzheimer's disease (AD), but their relative performance in predicting future cognitive decline among cognitively unimpaired (CU) individuals is unclear. In this head-to-head comparison study including 9 cohorts and 1534 individuals, we found that plasma p-tau217 and medial temporal lobe Tau-PET signal showed similar associations with cognitive decline on a global cognitive composite test (R2 PET=0.32 vs R2 PLASMA=0.32, pdifference=0.812) and with progression to mild cognitive impairment (Hazard ratio[HR]PET=1.56[1.43-1.70] vs HRPLASMA=1.63[1.50-1.77], pdifference=0.627). Combined plasma and PET models were superior to the single biomarker models (R2=0.36, p<0.01). Furthermore, sequential selection using plasma p-tau217 and then Tau-PET reduced the number of participants required for a clinical trial by 94%, compared to a 75% reduction when using plasma p-tau217 alone. We conclude that plasma p-tau217 and Tau-PET showed similar performance for predicting future cognitive decline in CU individuals, and their sequential use (i.e., plasma p-tau217 followed by Tau-PET in a subset with high plasma p-tau217) is useful for screening in clinical trials in preclinical AD.
Collapse
Affiliation(s)
- Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Divya Bali
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Linda Karlsson
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Emma M. Coomans
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Elsmarieke van de Giessen
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Wiesje M. van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Department of Epidemiology and Biostatistics, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Charlotte E. Teunissen
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Neurochemistry Laboratory, Department of Laboratory Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Erin M. Jonaitis
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison
| | - Sterling C. Johnson
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison
| | | | - Sylvia Villeneuve
- Douglas Mental Health University Institute, Centre for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Tammie L.S. Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington, Washington University School of Medicine, St. Louis, MO, USA
| | - Suzanne E. Schindler
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - Randall J. Bateman
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
- The Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO, United States
| | - James D. Doecke
- Australian eHealth Research Centre, CSIRO, Melbourne, Victoria, Australia
| | - Vincent Doré
- Australian eHealth Research Centre, CSIRO, Melbourne, Victoria, Australia
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, Australia
| | - Azadeh Feizpour
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Colin L. Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Christopher Rowe
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Heather J. Wiste
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
18
|
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
|
19
|
Chen Y, Al-Nusaif M, Li S, Tan X, Yang H, Cai H, Le W. Progress on early diagnosing Alzheimer's disease. Front Med 2024; 18:446-464. [PMID: 38769282 PMCID: PMC11391414 DOI: 10.1007/s11684-023-1047-1] [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: 09/23/2023] [Accepted: 11/15/2023] [Indexed: 05/22/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects both cognition and non-cognition functions. The disease follows a continuum, starting with preclinical stages, progressing to mild cognitive and behavioral impairment, ultimately leading to dementia. Early detection of AD is crucial for better diagnosis and more effective treatment. However, the current AD diagnostic tests of biomarkers using cerebrospinal fluid and/or brain imaging are invasive or expensive, and mostly are still not able to detect early disease state. Consequently, there is an urgent need to develop new diagnostic techniques with higher sensitivity and specificity during the preclinical stages of AD. Various non-cognitive manifestations, including behavioral abnormalities, sleep disturbances, sensory dysfunctions, and physical changes, have been observed in the preclinical AD stage before occurrence of notable cognitive decline. Recent research advances have identified several biofluid biomarkers as early indicators of AD. This review focuses on these non-cognitive changes and newly discovered biomarkers in AD, specifically addressing the preclinical stages of the disease. Furthermore, it is of importance to explore the potential for developing a predictive system or network to forecast disease onset and progression at the early stage of AD.
Collapse
Affiliation(s)
- Yixin Chen
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Murad Al-Nusaif
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Song Li
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Xiang Tan
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Huijia Yang
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China.
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China.
| |
Collapse
|
20
|
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
|
21
|
Zhou L, Butler TA, Wang XH, Xi K, Tanzi EB, Glodzik L, Chiang GC, de Leon MJ, Li Y. Multimodal assessment of brain fluid clearance is associated with amyloid-beta deposition in humans. J Neuroradiol 2024; 51:101164. [PMID: 37907155 PMCID: PMC11058119 DOI: 10.1016/j.neurad.2023.10.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/09/2023] [Accepted: 10/28/2023] [Indexed: 11/02/2023]
Abstract
PURPOSE The present study investigates a multimodal imaging assessment of glymphatic function and its association with brain amyloid-beta deposition. METHODS Two brain CSF clearance measures (vCSF and DTI-ALPS) were derived from dynamic PET and MR diffusion tensor imaging (DTI) for 50 subjects, 24/50 were Aβ positive (Aβ+). T1W, T2W, DTI, T2FLAIR, and 11C-PiB and 18F-MK-6240 PET were acquired. Multivariate linear regression models were assessed with both vCSF and DTI-ALPS as independent variables and brain Aβ as the dependent variable. Three types of models were evaluated, including the vCSF-only model, the ALPS-only model and the vCSF+ALPS combined model. Models were applied to the whole group, and Aβ subgroups. All analyses were controlled for age, gender, and intracranial volume. RESULTS Sample demographics (N=50) include 20 males and 30 females with a mean age of 69.30 (sd=8.55). Our results show that the combination of vCSF and ALPS associates with Aβ deposition (p < 0.05, R2 = 0.575) better than either vCSF (p < 0.05, R2 = 0.431) or ALPS (p < 0.05, R2 = 0.372) alone in the Aβ+ group. We observed similar results in whole-group analyses (combined model: p < 0.05, R2 = 0.287; vCSF model: p <0.05, R2 = 0.175; ALPS model: p < 0.05, R2 = 0.196) with less significance. Our data also showed that vCSF has higher correlation (r = -0.548) in subjects with mild Aβ deposition and DTI-ALPS has higher correlation (r=-0.451) with severe Aβ deposition subjects. CONCLUSION The regression model with both vCSF and DTI-ALPS is better associated with brain Aβ deposition. These two independent brain clearance measures may better explain the variation in Aβ deposition than either term individually. Our results suggest that vCSF and DTI-ALPS reflect complementary aspects of brain clearance functions.
Collapse
Affiliation(s)
- Liangdong Zhou
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 E 61st St, Feil 2, New York, NY 10065, United States
| | - Tracy A Butler
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 E 61st St, Feil 2, New York, NY 10065, United States
| | - Xiuyuan H Wang
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 E 61st St, Feil 2, New York, NY 10065, United States
| | - Ke Xi
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 E 61st St, Feil 2, New York, NY 10065, United States
| | - Emily B Tanzi
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 E 61st St, Feil 2, New York, NY 10065, United States
| | - Lidia Glodzik
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 E 61st St, Feil 2, New York, NY 10065, United States
| | - Gloria C Chiang
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 E 61st St, Feil 2, New York, NY 10065, United States
| | - Mony J de Leon
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 E 61st St, Feil 2, New York, NY 10065, United States
| | - Yi Li
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 E 61st St, Feil 2, New York, NY 10065, United States.
| |
Collapse
|
22
|
Sanchez-Rodriguez LM, Bezgin G, Carbonell F, Therriault J, Fernandez-Arias J, Servaes S, Rahmouni N, Tissot C, Stevenson J, Karikari TK, Ashton NJ, Benedet AL, Zetterberg H, Blennow K, Triana-Baltzer G, Kolb HC, Rosa-Neto P, Iturria-Medina Y. Personalized whole-brain neural mass models reveal combined Aβ and tau hyperexcitable influences in Alzheimer's disease. Commun Biol 2024; 7:528. [PMID: 38704445 PMCID: PMC11069569 DOI: 10.1038/s42003-024-06217-2] [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: 08/24/2023] [Accepted: 04/19/2024] [Indexed: 05/06/2024] Open
Abstract
Neuronal dysfunction and cognitive deterioration in Alzheimer's disease (AD) are likely caused by multiple pathophysiological factors. However, mechanistic evidence in humans remains scarce, requiring improved non-invasive techniques and integrative models. We introduce personalized AD computational models built on whole-brain Wilson-Cowan oscillators and incorporating resting-state functional MRI, amyloid-β (Aβ) and tau-PET from 132 individuals in the AD spectrum to evaluate the direct impact of toxic protein deposition on neuronal activity. This subject-specific approach uncovers key patho-mechanistic interactions, including synergistic Aβ and tau effects on cognitive impairment and neuronal excitability increases with disease progression. The data-derived neuronal excitability values strongly predict clinically relevant AD plasma biomarker concentrations (p-tau217, p-tau231, p-tau181, GFAP) and grey matter atrophy obtained through voxel-based morphometry. Furthermore, reconstructed EEG proxy quantities show the hallmark AD electrophysiological alterations (theta band activity enhancement and alpha reductions) which occur with Aβ-positivity and after limbic tau involvement. Microglial activation influences on neuronal activity are less definitive, potentially due to neuroimaging limitations in mapping neuroprotective vs detrimental activation phenotypes. Mechanistic brain activity models can further clarify intricate neurodegenerative processes and accelerate preventive/treatment interventions.
Collapse
Affiliation(s)
- Lazaro M Sanchez-Rodriguez
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, QC, Canada
| | - Gleb Bezgin
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | | | - Joseph Therriault
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Jaime Fernandez-Arias
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Stijn Servaes
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Nesrine Rahmouni
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Cécile Tissot
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
- Lawrence Berkeley National Laboratory, Berkeley, USA
| | - Jenna Stevenson
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- King's College London, Institute of Psychiatry, Psychology and Neuroscience Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Andréa L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | | | - Hartmuth C Kolb
- Neuroscience Biomarkers, Janssen Research & Development, La Jolla, CA, USA
| | - Pedro Rosa-Neto
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Yasser Iturria-Medina
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada.
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, QC, Canada.
| |
Collapse
|
23
|
Nordberg A. Insights into the progression of genetic Alzheimer's disease from tau PET. Lancet Neurol 2024; 23:453-454. [PMID: 38631755 DOI: 10.1016/s1474-4422(24)00124-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024]
Affiliation(s)
- Agneta Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Theme Inflammation and Aging, Karolinska University Hospital, Stockholm 14183, Sweden.
| |
Collapse
|
24
|
Salvadó G, Horie K, Barthélemy NR, Vogel JW, Pichet Binette A, Chen CD, Aschenbrenner AJ, Gordon BA, Benzinger TLS, Holtzman DM, Morris JC, Palmqvist S, Stomrud E, Janelidze S, Ossenkoppele R, Schindler SE, Bateman RJ, Hansson O. Disease staging of Alzheimer's disease using a CSF-based biomarker model. NATURE AGING 2024; 4:694-708. [PMID: 38514824 PMCID: PMC11108782 DOI: 10.1038/s43587-024-00599-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 02/20/2024] [Indexed: 03/23/2024]
Abstract
Biological staging of individuals with Alzheimer's disease (AD) may improve diagnostic and prognostic workup of dementia in clinical practice and the design of clinical trials. In this study, we used the Subtype and Stage Inference (SuStaIn) algorithm to establish a robust biological staging model for AD using cerebrospinal fluid (CSF) biomarkers. Our analysis involved 426 participants from BioFINDER-2 and was validated in 222 participants from the Knight Alzheimer Disease Research Center cohort. SuStaIn identified a singular biomarker sequence and revealed that five CSF biomarkers effectively constituted a reliable staging model (ordered: Aβ42/40, pT217/T217, pT205/T205, MTBR-tau243 and non-phosphorylated mid-region tau). The CSF stages (0-5) demonstrated a correlation with increased abnormalities in other AD-related biomarkers, such as Aβ-PET and tau-PET, and aligned with longitudinal biomarker changes reflective of AD progression. Higher CSF stages at baseline were associated with an elevated hazard ratio of clinical decline. This study highlights a common molecular pathway underlying AD pathophysiology across all patients, suggesting that a single CSF collection can accurately indicate the presence of AD pathologies and characterize the stage of disease progression. The proposed staging model has implications for enhancing diagnostic and prognostic assessments in both clinical practice and the design of clinical trials.
Collapse
Affiliation(s)
- Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.
| | - Kanta Horie
- Tracy Family Stable Isotope Labeling Quantitation (SILQ) Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Eisai, Inc., Nutley, NJ, USA
| | - Nicolas R Barthélemy
- Tracy Family Stable Isotope Labeling Quantitation (SILQ) Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jacob W Vogel
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Clinical Science, Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Charles D Chen
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew J Aschenbrenner
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J Bateman
- Tracy Family Stable Isotope Labeling Quantitation (SILQ) Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
25
|
Ishibashi K, Kurihara M, Toyohara J, Ishii K, Iwata A. Pitfalls of Amyloid-Beta PET: Comparisons With 18 F-MK-6240 and 18 F-THK5351 PET. Clin Nucl Med 2024; 49:319-321. [PMID: 38363815 DOI: 10.1097/rlu.0000000000005097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2024]
Abstract
ABSTRACT We present 3 patients as pitfalls of amyloid-beta (Aβ) PET, who underwent 11 C-PiB (Aβ), 18 F-MK-6240 (Alzheimer disease [AD]-tau), and 18 F-THK5351 (astrogliosis) PET examinations. Despite negligible or tiny Aβ pathology, patients 1 and 2 were diagnosed with AD as the cause of symptoms. Despite widespread Aβ pathology, patient 3 was not diagnosed with AD as the cause of symptoms. However, if we had only conducted Aβ PET, patients 1 and 2 might not have been diagnosed with AD, whereas patient 3 might have been diagnosed with AD. Hence, both Aβ and AD-tau assessments are necessary to relate clinical symptoms to AD pathology.
Collapse
Affiliation(s)
| | - Masanori Kurihara
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | | | | | - Atsushi Iwata
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| |
Collapse
|
26
|
Therriault J, Schindler SE, Salvadó G, Pascoal TA, Benedet AL, Ashton NJ, Karikari TK, Apostolova L, Murray ME, Verberk I, Vogel JW, La Joie R, Gauthier S, Teunissen C, Rabinovici GD, Zetterberg H, Bateman RJ, Scheltens P, Blennow K, Sperling R, Hansson O, Jack CR, Rosa-Neto P. Biomarker-based staging of Alzheimer disease: rationale and clinical applications. Nat Rev Neurol 2024; 20:232-244. [PMID: 38429551 DOI: 10.1038/s41582-024-00942-2] [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] [Accepted: 02/05/2024] [Indexed: 03/03/2024]
Abstract
Disease staging, whereby the spatial extent and load of brain pathology are used to estimate the severity of Alzheimer disease (AD), is pivotal to the gold-standard neuropathological diagnosis of AD. Current in vivo diagnostic frameworks for AD are based on abnormal concentrations of amyloid-β and tau in the cerebrospinal fluid or on PET scans, and breakthroughs in molecular imaging have opened up the possibility of in vivo staging of AD. Focusing on the key principles of disease staging shared across several areas of medicine, this Review highlights the potential for in vivo staging of AD to transform our understanding of preclinical AD, refine enrolment criteria for trials of disease-modifying therapies and aid clinical decision-making in the era of anti-amyloid therapeutics. We provide a state-of-the-art review of recent biomarker-based AD staging systems and highlight their contributions to the understanding of the natural history of AD. Furthermore, we outline hypothetical frameworks to stage AD severity using more accessible fluid biomarkers. In addition, by applying amyloid PET-based staging to recently published anti-amyloid therapeutic trials, we highlight how biomarker-based disease staging frameworks could illustrate the numerous pathological changes that have already taken place in individuals with mildly symptomatic AD. Finally, we discuss challenges related to the validation and standardization of disease staging and provide a forward-looking perspective on potential clinical applications.
Collapse
Affiliation(s)
- Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, Quebec, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada.
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andréa Lessa Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- NIHR Biomedical Research Centre, South London and Maudsley NHS Foundation, London, UK
| | - Thomas K Karikari
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Liana Apostolova
- Department of Neurology, University of Indiana School of Medicine, Indianapolis, IN, USA
| | | | - Inge Verberk
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Jacob W Vogel
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Clinical Sciences, Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Charlotte Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Philip Scheltens
- Alzheimer Centre Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Reisa Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | | | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
27
|
Vanderlinden G, Michiels L, Koole M, Lemmens R, Liessens D, Van Walleghem J, Depreitere B, Vandenbulcke M, Van Laere K. Tau Imaging in Late Traumatic Brain Injury: A [ 18F]MK-6240 Positron Emission Tomography Study. J Neurotrauma 2024; 41:420-429. [PMID: 38038357 DOI: 10.1089/neu.2023.0085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023] Open
Abstract
Epidemiological studies have identified prior traumatic brain injury (TBI) as a risk factor for developing Alzheimer's disease (AD). Neurofibrillary tangles (NFTs) are common to AD and chronic traumatic encephalopathy following repetitive mild TBI. However, it is unclear if a single TBI is sufficient to cause accumulation of NFTs. We performed a [18F]MK-6240 positron emission tomography (PET) imaging study to assess NFTs in patients who had sustained a single TBI at least 2 years prior to study inclusion. Fourteen TBI patients (49 ± 20 years; 5 M/9 F; 8 moderate-severe, 1 mild-probable, 5 symptomatic-possible TBI) and 40 demographically similar controls (57 ± 19 years; 19 M/21 F) underwent simultaneous [18F]MK-6240 PET and magnetic resonance imaging (MRI) as well as neuropsychological assessment including the Cambridge Neuropsychological Test Automated Battery (CANTAB). A region-based voxelwise partial volume correction was applied, using parcels obtained by FreeSurfer v6.0, and standardized uptake value ratios (SUVR) were calculated relative to the cerebellar gray matter. Group differences were assessed on both a voxel- and a volume-of-interest-based level and correlations of [18F]MK-6240 SUVR with time since injury as well as with clinical outcomes were calculated. Visual assessment of TBI images did not show global or focal increases in tracer uptake in any subject. On a group level, [18F]MK-6240 SUVR was not significantly different in patients versus controls or between subgroups of moderate-severe TBI versus less severe TBI. Within the TBI group, One Touch Stockings problem solving and spatial working memory (executive function), reaction time (attention), and Mini-Mental State Examination (MMSE) (global cognition) were associated with [18F]MK-6240 SUVR. We found no group-based increase of [18F]MK-6240 brain uptake in patients scanned at least 2 years after a single TBI compared with healthy volunteers, which suggests that no NFTs are building up in the first years after a single TBI. Nonetheless, correlations with cognitive outcomes were found that warrant further investigation.
Collapse
Affiliation(s)
- Greet Vanderlinden
- Nuclear Medicine and Molecular Imaging, Imaging and Pathology, and Departments of Research Group Psychiatry, KU Leuven, Leuven, Belgium
| | - Laura Michiels
- Leuven Brain Institute, Leuven, Belgium
- Department of Neurology, University Hospitals UZ Leuven, Leuven, Belgium
- VIB, Center for Brain and Disease Research, Laboratory of Neurobiology, Belgium
- Neurosciences, and Research Group Psychiatry, KU Leuven, Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, Imaging and Pathology, and Departments of Research Group Psychiatry, KU Leuven, Leuven, Belgium
| | - Robin Lemmens
- Leuven Brain Institute, Leuven, Belgium
- Department of Neurology, University Hospitals UZ Leuven, Leuven, Belgium
- VIB, Center for Brain and Disease Research, Laboratory of Neurobiology, Belgium
- Neurosciences, and Research Group Psychiatry, KU Leuven, Leuven, Belgium
| | - Dirk Liessens
- Department of Geriatric Psychiatry, University Hospitals UZ Leuven, Leuven, Belgium
| | | | - Bart Depreitere
- Department of Neurosurgery, and University Hospitals UZ Leuven, Leuven, Belgium
| | - Mathieu Vandenbulcke
- Leuven Brain Institute, Leuven, Belgium
- Department of Geriatric Psychiatry, University Hospitals UZ Leuven, Leuven, Belgium
- Neuropsychiatry, Research Group Psychiatry, KU Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Imaging and Pathology, and Departments of Research Group Psychiatry, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, Leuven, Belgium
- Department of Nuclear Medicine, University Hospitals UZ Leuven, Leuven, Belgium
| |
Collapse
|
28
|
Woo MS, Tissot C, Lantero‐Rodriguez J, Snellman A, Therriault J, Rahmouni N, Macedo AC, Servaes S, Wang Y, Arias JF, Hosseini SA, Chamoun M, Lussier FZ, Benedet AL, Ashton NJ, Karikari TK, Triana‐Baltzer G, Kolb HC, Stevenson J, Mayer C, Kobayashi E, Massarweh G, Friese MA, Pascoal TA, Gauthier S, Zetterberg H, Blennow K, Rosa‐Neto P. Plasma pTau-217 and N-terminal tau (NTA) enhance sensitivity to identify tau PET positivity in amyloid-β positive individuals. Alzheimers Dement 2024; 20:1166-1174. [PMID: 37920945 PMCID: PMC10916953 DOI: 10.1002/alz.13528] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 11/04/2023]
Abstract
INTRODUCTION We set out to identify tau PET-positive (A+T+) individuals among amyloid-beta (Aβ) positive participants using plasma biomarkers. METHODS In this cross-sectional study we assessed 234 participants across the AD continuum who were evaluated by amyloid PET with [18 F]AZD4694 and tau-PET with [18 F]MK6240 and measured plasma levels of total tau, pTau-181, pTau-217, pTau-231, and N-terminal tau (NTA-tau). We evaluated the performances of plasma biomarkers to predict tau positivity in Aβ+ individuals. RESULTS Highest associations with tau positivity in Aβ+ individuals were found for plasma pTau-217 (AUC [CI95% ] = 0.89 [0.82, 0.96]) and NTA-tau (AUC [CI95% ] = 0.88 [0.91, 0.95]). Combining pTau-217 and NTA-tau resulted in the strongest agreement (Cohen's Kappa = 0.74, CI95% = 0.57/0.90, sensitivity = 92%, specificity = 81%) with PET for classifying tau positivity. DISCUSSION The potential for identifying tau accumulation in later Braak stages will be useful for patient stratification and prognostication in treatment trials and in clinical practice. HIGHLIGHTS We found that in a cohort without pre-selection pTau-181, pTau-217, and NTA-tau showed the highest association with tau PET positivity. We found that in Aβ+ individuals pTau-217 and NTA-tau showed the highest association with tau PET positivity. Combining pTau-217 and NTA-tau resulted in the strongest agreement with the tau PET-based classification.
Collapse
|
29
|
Aguero C, Dhaynaut M, Amaral AC, Moon SH, Neelamegam R, Scapellato M, Carazo-Casas C, Kumar S, El Fakhri G, Johnson K, Frosch MP, Normandin MD, Gómez-Isla T. Head-to-head comparison of [ 18F]-Flortaucipir, [ 18F]-MK-6240 and [ 18F]-PI-2620 postmortem binding across the spectrum of neurodegenerative diseases. Acta Neuropathol 2024; 147:25. [PMID: 38280071 PMCID: PMC10822013 DOI: 10.1007/s00401-023-02672-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 01/29/2024]
Abstract
We and others have shown that [18F]-Flortaucipir, the most validated tau PET tracer thus far, binds with strong affinity to tau aggregates in Alzheimer's (AD) but has relatively low affinity for tau aggregates in non-AD tauopathies and exhibits off-target binding to neuromelanin- and melanin-containing cells, and to hemorrhages. Several second-generation tau tracers have been subsequently developed. [18F]-MK-6240 and [18F]-PI-2620 are the two that have garnered most attention. Our recent data indicated that the binding pattern of [18F]-MK-6240 closely parallels that of [18F]-Flortaucipir. The present study aimed at the direct comparison of the autoradiographic binding properties and off-target profile of [18F]-Flortaucipir, [18F]-MK-6240 and [18F]-PI-2620 in human tissue specimens, and their potential binding to monoamine oxidases (MAO). Phosphor-screen and high resolution autoradiographic patterns of the three tracers were studied in the same postmortem tissue material from AD and non-AD tauopathies, cerebral amyloid angiopathy, synucleopathies, transactive response DNA-binding protein 43 (TDP-43)-frontotemporal lobe degeneration and controls. Our results show that the three tracers show nearly identical autoradiographic binding profiles. They all strongly bind to neurofibrillary tangles in AD but do not seem to bind to a significant extent to tau aggregates in non-AD tauopathies pointing to their limited utility for the in vivo detection of non-AD tau lesions. None of them binds to lesions containing β-amyloid, α-synuclein or TDP-43 but they all show strong off-target binding to neuromelanin and melanin-containing cells, as well as weaker binding to areas of hemorrhage. The autoradiographic binding signals of the three tracers are only weakly displaced by competing concentrations of selective MAO-B inhibitor deprenyl but not by MAO-A inhibitor clorgyline suggesting that MAO enzymes do not appear to be a significant binding target of any of them. These findings provide relevant insights for the correct interpretation of the in vivo behavior of these three tau PET tracers.
Collapse
Affiliation(s)
- Cinthya Aguero
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Maeva Dhaynaut
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Ana C Amaral
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - S-H Moon
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Ramesh Neelamegam
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Margaret Scapellato
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Carlos Carazo-Casas
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Sunny Kumar
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Keith Johnson
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Matthew P Frosch
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| | - Marc D Normandin
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Teresa Gómez-Isla
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA.
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA.
| |
Collapse
|
30
|
Therriault J, Woo MS, Salvadó G, Gobom J, Karikari TK, Janelidze S, Servaes S, Rahmouni N, Tissot C, Ashton NJ, Benedet AL, Montoliu-Gaya L, Macedo AC, Lussier FZ, Stevenson J, Vitali P, Friese MA, Massarweh G, Soucy JP, Pascoal TA, Stomrud E, Palmqvist S, Mattsson-Carlgren N, Gauthier S, Zetterberg H, Hansson O, Blennow K, Rosa-Neto P. Comparison of immunoassay- with mass spectrometry-derived p-tau quantification for the detection of Alzheimer's disease pathology. Mol Neurodegener 2024; 19:2. [PMID: 38185677 PMCID: PMC10773025 DOI: 10.1186/s13024-023-00689-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 11/28/2023] [Indexed: 01/09/2024] Open
Abstract
BACKGROUND Antibody-based immunoassays have enabled quantification of very low concentrations of phosphorylated tau (p-tau) protein forms in cerebrospinal fluid (CSF), aiding in the diagnosis of AD. Mass spectrometry enables absolute quantification of multiple p-tau variants within a single run. The goal of this study was to compare the performance of mass spectrometry assessments of p-tau181, p-tau217 and p-tau231 with established immunoassay techniques. METHODS We measured p-tau181, p-tau217 and p-tau231 concentrations in CSF from 173 participants from the TRIAD cohort and 394 participants from the BioFINDER-2 cohort using both mass spectrometry and immunoassay methods. All subjects were clinically evaluated by dementia specialists and had amyloid-PET and tau-PET assessments. Bland-Altman analyses evaluated the agreement between immunoassay and mass spectrometry p-tau181, p-tau217 and p-tau231. P-tau associations with amyloid-PET and tau-PET uptake were also compared. Receiver Operating Characteristic (ROC) analyses compared the performance of mass spectrometry and immunoassays p-tau concentrations to identify amyloid-PET positivity. RESULTS Mass spectrometry and immunoassays of p-tau217 were highly comparable in terms of diagnostic performance, between-group effect sizes and associations with PET biomarkers. In contrast, p-tau181 and p-tau231 concentrations measured using antibody-free mass spectrometry had lower performance compared with immunoassays. CONCLUSIONS Our results suggest that while similar overall, immunoassay-based p-tau biomarkers are slightly superior to antibody-free mass spectrometry-based p-tau biomarkers. Future work is needed to determine whether the potential to evaluate multiple biomarkers within a single run offsets the slightly lower performance of antibody-free mass spectrometry-based p-tau quantification.
Collapse
Affiliation(s)
- Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Marcel S Woo
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada
- Department of Neurology, Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, 20251, Germany
| | - Gemma Salvadó
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, S-431 80, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, S-431 80, Sweden
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, S-431 80, Sweden
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, 15213, USA
| | - Shorena Janelidze
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, S-431 80, Sweden
- Wallenberg Centre for Molecular Medicine, University of Gothenburg, Gothenburg, S-413 45, Sweden
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, SE5 9RT, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, SE5 8AF, UK
| | - Andréa Lessa Benedet
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, S-431 80, Sweden
| | - Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, S-431 80, Sweden
| | - Arthur C Macedo
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Firoza Z Lussier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, 15213, USA
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada
| | - Paolo Vitali
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Manuel A Friese
- Department of Neurology, Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, 20251, Germany
| | - Gassan Massarweh
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Jean-Paul Soucy
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, 15213, USA
| | - Erik Stomrud
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Sebastian Palmqvist
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, S-431 80, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, S-431 80, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1N 6BG, UK
- UK Dementia Research Institute at UCL, London, WC1N 6BG, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Oskar Hansson
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, S-431 80, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, S-431 80, Sweden
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada.
| |
Collapse
|
31
|
Lantero-Rodriguez J, Montoliu-Gaya L, Benedet AL, Vrillon A, Dumurgier J, Cognat E, Brum WS, Rahmouni N, Stevenson J, Servaes S, Therriault J, Becker B, Brinkmalm G, Snellman A, Huber H, Kvartsberg H, Ashton NJ, Zetterberg H, Paquet C, Rosa-Neto P, Blennow K. CSF p-tau205: a biomarker of tau pathology in Alzheimer's disease. Acta Neuropathol 2024; 147:12. [PMID: 38184490 PMCID: PMC10771353 DOI: 10.1007/s00401-023-02659-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/31/2023] [Accepted: 11/16/2023] [Indexed: 01/08/2024]
Abstract
Post-mortem staging of Alzheimer's disease (AD) neurofibrillary pathology is commonly performed by immunohistochemistry using AT8 antibody for phosphorylated tau (p-tau) at positions 202/205. Thus, quantification of p-tau205 and p-tau202 in cerebrospinal fluid (CSF) should be more reflective of neurofibrillary tangles in AD than other p-tau epitopes. We developed two novel Simoa immunoassays for CSF p-tau205 and p-tau202 and measured these phosphorylations in three independent cohorts encompassing the AD continuum, non-AD cases and cognitively unimpaired participants: a discovery cohort (n = 47), an unselected clinical cohort (n = 212) and a research cohort well-characterized by fluid and imaging biomarkers (n = 262). CSF p-tau205 increased progressively across the AD continuum, while CSF p-tau202 was increased only in AD and amyloid (Aβ) and tau pathology positive (A+T+) cases (P < 0.01). In A+ cases, CSF p-tau205 and p-tau202 showed stronger associations with tau-PET (rSp205 = 0.67, rSp202 = 0.45) than Aβ-PET (rSp205 = 0.40, rSp202 = 0.09). CSF p-tau205 increased gradually across tau-PET Braak stages (P < 0.01), whereas p-tau202 only increased in Braak V-VI (P < 0.0001). Both showed stronger regional associations with tau-PET than with Aβ-PET, and CSF p-tau205 was significantly associated with Braak V-VI tau-PET regions. When assessing the contribution of Aβ and tau pathologies (indexed by PET) to CSF p-tau205 and p-tau202 variance, tau pathology was found to be the most prominent contributor in both cases (CSF p-tau205: R2 = 69.7%; CSF p-tau202: R2 = 85.6%) Both biomarkers associated with brain atrophy measurements globally (rSp205 = - 0.36, rSp202 = - 0.33) and regionally, and correlated with cognition (rSp205 = - 0.38/- 0.40, rSp202 = - 0.20/- 0.29). In conclusion, we report the first high-throughput CSF p-tau205 immunoassay for the in vivo quantification of tau pathology in AD, and a potentially cost-effective alternative to tau-PET in clinical settings and clinical trials.
Collapse
Affiliation(s)
- Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
| | - Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Andrea L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Agathe Vrillon
- Cognitive Neurology Center, Université de Paris Cité, GHU Nord APHP Hospital Lariboisière Fernand Widal, Paris, France
| | - Julien Dumurgier
- Cognitive Neurology Center, Université de Paris Cité, GHU Nord APHP Hospital Lariboisière Fernand Widal, Paris, France
| | - Emmanuel Cognat
- Cognitive Neurology Center, Université de Paris Cité, GHU Nord APHP Hospital Lariboisière Fernand Widal, Paris, France
| | - Wagner S Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil
| | - Nesrine Rahmouni
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Jenna Stevenson
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Stijn Servaes
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Joseph Therriault
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Bruno Becker
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Gunnar Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Anniina Snellman
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
| | - Hanna Huber
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Hlin Kvartsberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| | - Claire Paquet
- Cognitive Neurology Center, Université de Paris Cité, GHU Nord APHP Hospital Lariboisière Fernand Widal, Paris, France
| | - Pedro Rosa-Neto
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
32
|
Wang Y, Zhang Y, Yu E. Targeted examination of amyloid beta and tau protein accumulation via positron emission tomography for the differential diagnosis of Alzheimer's disease based on the A/T(N) research framework. Clin Neurol Neurosurg 2024; 236:108071. [PMID: 38043158 DOI: 10.1016/j.clineuro.2023.108071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 11/24/2023] [Accepted: 11/25/2023] [Indexed: 12/05/2023]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases among the older population. Its main pathological features include the abnormal deposition of extracellular amyloid-β plaques and the intracellular neurofibrillary tangles of tau proteins. Its clinical presentation is complex. This review introduces the pathological processes in AD and other common neurodegenerative diseases. It then discusses the positron emission tomography (PET) probes that target amyloid-β plaques and tau proteins for diagnosing AD. According to the A/T(N) research framework, combined targeted amyloid-β and tau protein detection via PET to further improve the diagnostic accuracy of AD. In particular, the properties of the 18F-flortaucipir and 18F-MK6240 tracers-may be more beneficial in helping to differentiate AD from other common neurodegenerative diseases, such as dementia with Lewy bodies, Parkinson's disease dementia, and frontotemporal dementia. Furthermore, the A/T(N) research framework should be used as the clinical diagnosis model of AD in the future.
Collapse
Affiliation(s)
- Ye Wang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Psychiatry, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), 310022, China
| | - Yuhan Zhang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Enyan Yu
- Department of Psychiatry, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), 310022, China.
| |
Collapse
|
33
|
Zhang M, Mi N, Ying Z, Lin X, Jin Y. Advances in the prevention and treatment of Alzheimer's disease based on oral bacteria. Front Psychiatry 2023; 14:1291455. [PMID: 38156323 PMCID: PMC10754487 DOI: 10.3389/fpsyt.2023.1291455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 11/22/2023] [Indexed: 12/30/2023] Open
Abstract
With the global population undergoing demographic shift towards aging, the prevalence of Alzheimer's disease (AD), a prominent neurodegenerative disorder that primarily afflicts individuals aged 65 and above, has increased across various geographical regions. This phenomenon is accompanied by a concomitant decline in immune functionality and oral hygiene capacity among the elderly, precipitating compromised oral functionality and an augmented burden of dental plaque. Accordingly, oral afflictions, including dental caries and periodontal disease, manifest with frequency among the geriatric population worldwide. Recent scientific investigations have unveiled the potential role of oral bacteria in instigating both local and systemic chronic inflammation, thereby delineating a putative nexus between oral health and the genesis and progression of AD. They further proposed the oral microbiome as a potentially modifiable risk factor in AD development, although the precise pathological mechanisms and degree of association have yet to be fully elucidated. This review summarizes current research on the relationship between oral bacteria and AD, describing the epidemiological and pathological mechanisms that may potentially link them. The purpose is to enrich early diagnostic approaches by incorporating emerging biomarkers, offering novel insights for clinicians in the early detection of AD. Additionally, it explores the potential of vaccination strategies and guidance for clinical pharmacotherapy. It proposes the development of maintenance measures specifically targeting oral health in older adults and advocates for guiding elderly patients in adopting healthy lifestyle habits, ultimately aiming to indirectly mitigate the progression of AD while promoting oral health in the elderly.
Collapse
Affiliation(s)
| | | | | | | | - Ying Jin
- Department of Stomatology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
34
|
Lantero-Rodriguez J, Tissot C, Snellman A, Servaes S, Benedet AL, Rahmouni N, Montoliu-Gaya L, Therriault J, Brum WS, Stevenson J, Lussier FZ, Bezgin G, Macedo AC, Chamoun M, Mathotaarachi SS, Pascoal TA, Ashton NJ, Zetterberg H, Neto PR, Blennow K. Plasma and CSF concentrations of N-terminal tau fragments associate with in vivo neurofibrillary tangle burden. Alzheimers Dement 2023; 19:5343-5354. [PMID: 37190913 DOI: 10.1002/alz.13119] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/16/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023]
Abstract
INTRODUCTION Fluid biomarkers capable of specifically tracking tau tangle pathology in vivo are greatly needed. METHODS We measured cerebrospinal fluid (CSF) and plasma concentrations of N-terminal tau fragments (NTA-tau), using a novel immunoassay (NTA) in the TRIAD cohort, consisting of 272 individuals assessed with amyloid beta (Aβ) positron emission tomography (PET), tau PET, magnetic resonance imaging (MRI) and cognitive assessments. RESULTS CSF and plasma NTA-tau concentrations were specifically increased in cognitively impaired Aβ-positive groups. CSF and plasma NTA-tau concentrations displayed stronger correlations with tau PET than with Aβ PET and MRI, both in global uptake and at the voxel level. Regression models demonstrated that both CSF and plasma NTA-tau are preferentially associated with tau pathology. Moreover, plasma NTA-tau was associated with longitudinal tau PET accumulation across the aging and Alzheimer's disease (AD) spectrum. DISCUSSION NTA-tau is a biomarker closely associated with in vivo tau deposition in the AD continuum and has potential as a tau tangle biomarker in clinical settings and trials. HIGHLIGHTS An assay for detecting N-terminal tau fragments (NTA-tau) in plasma and CSF was evaluated. NTA-tau is more closely associated with tau PET than amyloid PET or neurodegeneration. NTA-tau can successfully track in vivo tau deposition across the AD continuum. Plasma NTA-tau increased over time only in cognitively impaired amyloid-β positive individuals.
Collapse
Affiliation(s)
- Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Anniina Snellman
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Andrea L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Wagner S Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Firoza Z Lussier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Arthur C Macedo
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Sulantha S Mathotaarachi
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Pedro Rosa Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
35
|
Blazhenets G, Soleimani-Meigooni DN, Thomas W, Mundada N, Brendel M, Vento S, VandeVrede L, Heuer HW, Ljubenkov P, Rojas JC, Chen MK, Amuiri AN, Miller Z, Gorno-Tempini ML, Miller BL, Rosen HJ, Litvan I, Grossman M, Boeve B, Pantelyat A, Tartaglia MC, Irwin DJ, Dickerson BC, Baker SL, Boxer AL, Rabinovici GD, La Joie R. [ 18F]PI-2620 Binding Patterns in Patients with Suspected Alzheimer Disease and Frontotemporal Lobar Degeneration. J Nucl Med 2023; 64:1980-1989. [PMID: 37918868 PMCID: PMC10690126 DOI: 10.2967/jnumed.123.265856] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/27/2023] [Indexed: 11/04/2023] Open
Abstract
Tau PET has enabled the visualization of paired helical filaments of 3 or 4 C-terminal repeat tau in Alzheimer disease (AD), but its ability to detect aggregated tau in frontotemporal lobar degeneration (FTLD) spectrum disorders is uncertain. We investigated 2-(2-([18F]fluoro)pyridin-4-yl)-9H-pyrrolo[2,3-b:4,5c']dipyridine ([18F]PI-2620), a newer tracer with ex vivo evidence for binding to FTLD tau, in a convenience sample of patients with suspected FTLD and AD using a static acquisition protocol and parametric SUV ratio (SUVr) images. Methods: We analyzed [18F]PI-2620 PET data from 65 patients with clinical diagnoses associated with AD or FTLD neuropathology; most (60/65) also had amyloid-β (Aβ) PET. Scans were acquired 30-60 min after injection; SUVr maps (reference, inferior cerebellar cortex) were created for the full acquisition and for 10-min truncated sliding windows (30-40, 35-45,…50-60 min). Age- and sex-adjusted z score maps were computed for each patient, relative to 23 Aβ-negative cognitively healthy controls (HC). Mean SUVr in the globus pallidus, substantia nigra, subthalamic nuclei, dentate nuclei, white matter, and temporal gray matter was extracted for the full and truncated windows. Results: Patients with suspected AD neuropathology (Aβ-positive patients with mild cognitive impairment or AD dementia) showed high-intensity temporoparietal cortex-predominant [18F]PI-2620 binding. At the group level, patients with clinical diagnoses associated with FTLD (progressive supranuclear palsy with Richardson syndrome [PSP Richardson syndrome], corticobasal syndrome, and nonfluent-variant primary progressive aphasia) exhibited higher globus pallidus SUVr than did HCs; pallidal retention was highest in the PSP Richardson syndrome group, in whom SUVr was correlated with symptom severity (ρ = 0.53, P = 0.05). At the individual level, only half of PSP Richardson syndrome, corticobasal syndrome, and nonfluent-variant primary progressive aphasia patients had a pallidal SUVr above that of HCs. Temporal SUVr discriminated AD patients from HCs with high accuracy (area under the receiver operating characteristic curve, 0.94 [95% CI, 0.83-1.00]) for all time windows, whereas discrimination between patients with PSP Richardson syndrome and HCs using pallidal SUVr was fair regardless of time window (area under the receiver operating characteristic curve, 0.77 [95% CI, 0.61-0.92] at 30-40 min vs. 0.81 [95% CI, 0.66-0.96] at 50-60 min; P = 0.67). Conclusion: [18F]PI-2620 SUVr shows an intense and consistent signal in AD but lower-intensity, heterogeneous, and rapidly decreasing binding in patients with suspected FTLD. Further work is needed to delineate the substrate of [18F]PI-2620 binding and the usefulness of [18F]PI2620 SUVr quantification outside the AD continuum.
Collapse
Affiliation(s)
- Ganna Blazhenets
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
- Department of Nuclear Medicine, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - David N Soleimani-Meigooni
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Wesley Thomas
- Lawrence Berkeley National Laboratory, Berkeley, California
| | - Nidhi Mundada
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Stephanie Vento
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Lawren VandeVrede
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Hilary W Heuer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Peter Ljubenkov
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Julio C Rojas
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California
| | - Miranda K Chen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Alinda N Amuiri
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Zachary Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Maria L Gorno-Tempini
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Howie J Rosen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Irene Litvan
- University of California, San Diego, San Diego, California
| | - Murray Grossman
- Penn FTD Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | - David J Irwin
- Penn FTD Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California;
| |
Collapse
|
36
|
Woo MS, Nilsson J, Therriault J, Rahmouni N, Brinkmalm A, Benedet AL, Ashton NJ, Macedo AC, Servaes S, Wang YT, Tissot C, Arias JF, Hosseini SA, Chamoun M, Lussier FZ, Karikari TK, Stevenson J, Mayer C, Ferrari-Souza JP, Kobayashi E, Massarweh G, Friese MA, Pascoal TA, Gauthier S, Zetterberg H, Blennow K, Rosa-Neto P. 14-3-3 [Formula: see text]-reported early synaptic injury in Alzheimer's disease is independently mediated by sTREM2. J Neuroinflammation 2023; 20:278. [PMID: 38001539 PMCID: PMC10675887 DOI: 10.1186/s12974-023-02962-z] [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: 10/15/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
INTRODUCTION Synaptic loss is closely associated with tau aggregation and microglia activation in later stages of Alzheimer's disease (AD). However, synaptic damage happens early in AD at the very early stages of tau accumulation. It remains unclear whether microglia activation independently causes synaptic cleavage before tau aggregation appears. METHODS We investigated 104 participants across the AD continuum by measuring 14-3-3 zeta/delta ([Formula: see text]) as a cerebrospinal fluid biomarker for synaptic degradation, and fluid and imaging biomarkers of tau, amyloidosis, astrogliosis, neurodegeneration, and inflammation. We performed correlation analyses in cognitively unimpaired and impaired participants and used structural equation models to estimate the impact of microglia activation on synaptic injury in different disease stages. RESULTS 14-3-3 [Formula: see text] was increased in participants with amyloid pathology at the early stages of tau aggregation before hippocampal volume loss was detectable. 14-3-3 [Formula: see text] correlated with amyloidosis and tau load in all participants but only with biomarkers of neurodegeneration and memory deficits in cognitively unimpaired participants. This early synaptic damage was independently mediated by sTREM2. At later disease stages, tau and astrogliosis additionally mediated synaptic loss. CONCLUSIONS Our results advertise that sTREM2 is mediating synaptic injury at the early stages of tau accumulation, underlining the importance of microglia activation for AD disease propagation.
Collapse
Affiliation(s)
- Marcel S Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Johanna Nilsson
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 40530 Gothenburg, Sweden
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, 6875 La Salle Blvd, FBC Room 3149, Montreal, QC H4H 1R3 Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H4H 1R3 Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, 6875 La Salle Blvd, FBC Room 3149, Montreal, QC H4H 1R3 Canada
| | - Ann Brinkmalm
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 40530 Gothenburg, Sweden
| | - Andrea L Benedet
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, 40530 Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, 40530 Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Arthur C Macedo
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, 6875 La Salle Blvd, FBC Room 3149, Montreal, QC H4H 1R3 Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H4H 1R3 Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, 6875 La Salle Blvd, FBC Room 3149, Montreal, QC H4H 1R3 Canada
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, 6875 La Salle Blvd, FBC Room 3149, Montreal, QC H4H 1R3 Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H4H 1R3 Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, 6875 La Salle Blvd, FBC Room 3149, Montreal, QC H4H 1R3 Canada
| | - Jaime Fernandez Arias
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, 6875 La Salle Blvd, FBC Room 3149, Montreal, QC H4H 1R3 Canada
| | - Seyyed Ali Hosseini
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, 6875 La Salle Blvd, FBC Room 3149, Montreal, QC H4H 1R3 Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, 6875 La Salle Blvd, FBC Room 3149, Montreal, QC H4H 1R3 Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H4H 1R3 Canada
| | - Firoza Z Lussier
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H4H 1R3 Canada
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, 40530 Mölndal, Sweden
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, 6875 La Salle Blvd, FBC Room 3149, Montreal, QC H4H 1R3 Canada
| | - Christina Mayer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - João Pedro Ferrari-Souza
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 PA USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 91501-970 Brazil
| | - Eliane Kobayashi
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H4H 1R3 Canada
| | - Gassan Massarweh
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H4H 1R3 Canada
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Tharick A Pascoal
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 PA USA
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, 6875 La Salle Blvd, FBC Room 3149, Montreal, QC H4H 1R3 Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H4H 1R3 Canada
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 40530 Gothenburg, Sweden
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, 40530 Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1E 6BT UK
- UK Dementia Research Institute at UCL, London, WC1E 6BT UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, 518172 China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726 USA
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 40530 Gothenburg, Sweden
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, 40530 Mölndal, Sweden
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, 6875 La Salle Blvd, FBC Room 3149, Montreal, QC H4H 1R3 Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H4H 1R3 Canada
| |
Collapse
|
37
|
Aumont E, Bussy A, Bedard MA, Bezgin G, Therriault J, Savard M, Fernandez Arias J, Sziklas V, Vitali P, Poltronetti NM, Pallen V, Thomas E, Gauthier S, Kobayashi E, Rahmouni N, Stevenson J, Tissot C, Chakravarty MM, Rosa-Neto P. Hippocampal subfield associations with memory depend on stimulus modality and retrieval mode. Brain Commun 2023; 5:fcad309. [PMID: 38035364 PMCID: PMC10681971 DOI: 10.1093/braincomms/fcad309] [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: 05/24/2023] [Revised: 09/26/2023] [Accepted: 11/08/2023] [Indexed: 12/02/2023] Open
Abstract
Hippocampal atrophy is a well-known feature of age-related memory decline, and hippocampal subfields may contribute differently to this decline. In this cross-sectional study, we investigated the associations between hippocampal subfield volumes and performance in free recall and recognition memory tasks in both verbal and visual modalities in older adults without dementia. We collected MRIs from 97 (41 males) right-handed participants aged over 60. We segmented the right and left hippocampi into (i) dentate gyrus and cornu ammonis 4 (DG/CA4); (ii) CA2 and CA3 (CA2/CA3); (iii) CA1; (iv) strata radiatum, lacunosum and moleculare; and (v) subiculum. Memory was assessed with verbal free recall and recognition tasks, as well as visual free recall and recognition tasks. Amyloid-β and hippocampal tau positivity were assessed using [18F]AZD4694 and [18F]MK6240 PET tracers, respectively. The verbal free recall and verbal recognition performances were positively associated with CA1 and strata radiatum, lacunosum and moleculare volumes. The verbal free recall and visual free recall were positively correlated with the right DG/CA4. The visual free recall, but not verbal free recall, was also associated with the right CA2/CA3. The visual recognition was not significantly associated with any subfield volume. Hippocampal tau positivity, but not amyloid-β positivity, was associated with reduced DG/CA4, CA2/CA3 and strata radiatum, lacunosum and moleculare volumes. Our results suggest that memory performances are linked to specific subfields. CA1 appears to contribute to the verbal modality, irrespective of the free recall or recognition mode of retrieval. In contrast, DG/CA4 seems to be involved in the free recall mode, irrespective of verbal or visual modalities. These results are concordant with the view that DG/CA4 plays a primary role in encoding a stimulus' distinctive attributes, and that CA2/CA3 could be instrumental in recollecting a visual memory from one of its fragments. Overall, we show that hippocampal subfield segmentation can be useful for detecting early volume changes and improve our understanding of the hippocampal subfields' roles in memory.
Collapse
Affiliation(s)
- Etienne Aumont
- NeuroQAM Research Centre, Université du Québec à Montréal (UQAM), Montreal H2X 3P2, Canada
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Aurélie Bussy
- Cerebral Imaging Center, Douglas Research Center, Montreal, QC H4H 1R3, Canada
- Computational Brain Anatomy (CoBrALab) Laboratory, Montreal, QC H4H 1R2, Canada
| | - Marc-André Bedard
- NeuroQAM Research Centre, Université du Québec à Montréal (UQAM), Montreal H2X 3P2, Canada
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Gleb Bezgin
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Joseph Therriault
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Melissa Savard
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Jaime Fernandez Arias
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Viviane Sziklas
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Paolo Vitali
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | | | - Vanessa Pallen
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
| | - Emilie Thomas
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
| | - Serge Gauthier
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Eliane Kobayashi
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Nesrine Rahmouni
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Jenna Stevenson
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Cecile Tissot
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Mallar M Chakravarty
- Cerebral Imaging Center, Douglas Research Center, Montreal, QC H4H 1R3, Canada
- Computational Brain Anatomy (CoBrALab) Laboratory, Montreal, QC H4H 1R2, Canada
- Department of Psychiatry, McGill University, Montreal, QC H3A 1A1, Canada
| | - Pedro Rosa-Neto
- NeuroQAM Research Centre, Université du Québec à Montréal (UQAM), Montreal H2X 3P2, Canada
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| |
Collapse
|
38
|
Therriault J, Servaes S, Tissot C, Rahmouni N, Ashton NJ, Benedet AL, Karikari TK, Macedo AC, Lussier FZ, Stevenson J, Wang YT, Fernandez-Arias J, Stevenson A, Socualaya KQ, Haeger A, Nazneen T, Aumont É, Hosseini A, Rej S, Vitali P, Triana-Baltzer G, Kolb HC, Soucy JP, Pascoal TA, Gauthier S, Zetterberg H, Blennow K, Rosa-Neto P. Equivalence of plasma p-tau217 with cerebrospinal fluid in the diagnosis of Alzheimer's disease. Alzheimers Dement 2023; 19:4967-4977. [PMID: 37078495 PMCID: PMC10587362 DOI: 10.1002/alz.13026] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 04/21/2023]
Abstract
INTRODUCTION Plasma biomarkers are promising tools for Alzheimer's disease (AD) diagnosis, but comparisons with more established biomarkers are needed. METHODS We assessed the diagnostic performance of p-tau181 , p-tau217 , and p-tau231 in plasma and CSF in 174 individuals evaluated by dementia specialists and assessed with amyloid-PET and tau-PET. Receiver operating characteristic (ROC) analyses assessed the performance of plasma and CSF biomarkers to identify amyloid-PET and tau-PET positivity. RESULTS Plasma p-tau biomarkers had lower dynamic ranges and effect sizes compared to CSF p-tau. Plasma p-tau181 (AUC = 76%) and p-tau231 (AUC = 82%) assessments performed inferior to CSF p-tau181 (AUC = 87%) and p-tau231 (AUC = 95%) for amyloid-PET positivity. However, plasma p-tau217 (AUC = 91%) had diagnostic performance indistinguishable from CSF (AUC = 94%) for amyloid-PET positivity. DISCUSSION Plasma and CSF p-tau217 had equivalent diagnostic performance for biomarker-defined AD. Our results suggest that plasma p-tau217 may help reduce the need for invasive lumbar punctures without compromising accuracy in the identification of AD. HIGHLIGHTS p-tau217 in plasma performed equivalent to p-tau217 in CSF for the diagnosis of AD, suggesting the increased accessibility of plasma p-tau217 is not offset by lower accuracy. p-tau biomarkers in plasma had lower mean fold-changes between amyloid-PET negative and positive groups than p-tau biomarkers in CSF. CSF p-tau biomarkers had greater effect sizes than plasma p-tau biomarkers when differentiating between amyloid-PET positive and negative groups. Plasma p-tau181 and plasma p-tau231 performed worse than p-tau181 and p-tau231 in CSF for AD diagnosis.
Collapse
Affiliation(s)
- Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montréal, Québec, Canada, H4H 1R3
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada, H3A 2B4
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montréal, Québec, Canada, H4H 1R3
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada, H3A 2B4
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montréal, Québec, Canada, H4H 1R3
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montréal, Québec, Canada, H4H 1R3
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden, 6 431 41
- Wallenberg Centre for Molecular Medicine, University of Gothenburg, Gothenburg, Sweden, 6, 431 41
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, United Kingdom London, UK, SE5 9RT
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK, SE5 8AF
| | - Andréa Lessa Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden, 6 431 41
| | - Thomas K. Karikari
- Wallenberg Centre for Molecular Medicine, University of Gothenburg, Gothenburg, Sweden, 6, 431 41
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA, 15213
| | - Arthur C. Macedo
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montréal, Québec, Canada, H4H 1R3
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada, H3A 2B4
| | - Firoza Z. Lussier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montréal, Québec, Canada, H4H 1R3
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA, 15213
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montréal, Québec, Canada, H4H 1R3
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montréal, Québec, Canada, H4H 1R3
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada, H3A 2B4
| | - Jaime Fernandez-Arias
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montréal, Québec, Canada, H4H 1R3
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada, H3A 2B4
| | - Alyssa Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montréal, Québec, Canada, H4H 1R3
| | - Kely Quispialaya Socualaya
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montréal, Québec, Canada, H4H 1R3
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada, H3A 2B4
| | - Arlette Haeger
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montréal, Québec, Canada, H4H 1R3
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada, H3A 2B4
| | - Tahnia Nazneen
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montréal, Québec, Canada, H4H 1R3
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada, H3A 2B4
| | - Étienne Aumont
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montréal, Québec, Canada, H4H 1R3
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada, H3A 2B4
| | - Ali Hosseini
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montréal, Québec, Canada, H4H 1R3
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada, H3A 2B4
| | - Soham Rej
- Department of Psychiatry, McGill University Montreal, Quebec, Canada, H3T 1E2
| | - Paolo Vitali
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada, H3A 2B4
| | - Gallen Triana-Baltzer
- Neuroscience Biomarkers, Janssen Research & Development, La Jolla, California, USA, 92121
| | - Hartmuth C. Kolb
- Neuroscience Biomarkers, Janssen Research & Development, La Jolla, California, USA, 92121
| | - Jean-Paul Soucy
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada, H3A 2B4
| | - Tharick A. Pascoal
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA, 15213
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montréal, Québec, Canada, H4H 1R3
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada, H3A 2B4
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden, 6 431 41
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden, 6, 431 41
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK, SE5 9RT
- UK Dementia Research Institute at UCL, London, UK, SE5 9RT
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China, 0
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden, 6 431 41
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden, 6, 431 41
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montréal, Québec, Canada, H4H 1R3
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada, H3A 2B4
| |
Collapse
|
39
|
Chen SD, Zhang W, Feng YW, Wu BS, Yang L, Zhang YR, Wang HF, Guo Y, Deng YT, Feng JF, Cheng W, Dong Q, Yu JT. Genome-wide Survival Study Identifies PARL as a Novel Locus for Clinical Progression and Neurodegeneration in Alzheimer's Disease. Biol Psychiatry 2023; 94:732-742. [PMID: 36870520 DOI: 10.1016/j.biopsych.2023.02.992] [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: 09/02/2022] [Revised: 01/05/2023] [Accepted: 02/22/2023] [Indexed: 03/06/2023]
Abstract
BACKGROUND Variability exists in the trajectories of Alzheimer's disease (AD). We aimed to identify genetic modulators of clinical progression in AD. METHODS We conducted the first genome-wide survival study on AD using a two-stage approach. The discovery and replication stage separately included 1158 and 211,817 individuals without dementia from the Alzheimer's Disease Neuroimaging Initiative and the UK Biobank, respectively (325 and 1103 progressed in average follow-up of 4.33 and 8.63 years, respectively). Cox proportional hazards models were applied with time to AD dementia as the phenotype of clinical progression. A series of bioinformatic analyses and functional experiments was performed to validate the novel findings. RESULTS We found that APOE and PARL, a novel locus tagged by rs6795172 (hazard ratio = 1.66, p = 1.45 × 10-9), were significantly associated with AD clinical progression and were successfully replicated. The novel locus was linked to accelerated cognitive changes, higher tau levels, and faster atrophy of AD-specific brain structures, which were also verified in UK Biobank neuroimaging follow-up. Gene analysis and summary data-based Mendelian randomization indicated PARL as the most functionally relevant gene in the locus. Expression quantitative trait locus analyses and dual-luciferase reporter assays confirmed that PARL expression could be regulated by rs6795172. Three different AD mouse models consistently showed decreased PARL expression accompanied by elevated tau levels, and in vitro experiments revealed that knockdown/overexpression of PARL inversely changed tau levels. CONCLUSIONS Collectively, genetic, bioinformatic, and functional evidence suggests that PARL modulates clinical progression and neurodegeneration in AD. Targeting PARL may potentially modify AD progression and have implications for disease-modifying therapies.
Collapse
Affiliation(s)
- Shi-Dong Chen
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Wei Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, Shanghai, China
| | - Yi-Wei Feng
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Bang-Sheng Wu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Liu Yang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Hui-Fu Wang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, Shanghai, China
| | - Yu Guo
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Yue-Ting Deng
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China; Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, China; MOE Frontiers Center for Brain Science, Fudan University, Shanghai, Shanghai, China; Zhangjiang Fudan International Innovation Center, Shanghai, China
| | - Wei Cheng
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China; Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China; Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China.
| |
Collapse
|
40
|
Schaffer Aguzzoli C, Ferreira PCL, Povala G, Ferrari-Souza JP, Bellaver B, Soares Katz C, Zalzale H, Lussier FZ, Rohden F, Abbas S, Leffa DT, Scop Medeiros M, Therriault J, Benedet AL, Tissot C, Servaes S, Rahmouni N, Cassa Macedo A, Bezgin G, Kang MS, Stevenson J, Pallen V, Cohen A, Lopez OL, Tudorascu DL, Klunk WE, Villemagne VL, Soucy JP, Zimmer ER, Schilling LP, Karikari TK, Ashton NJ, Zetterberg H, Blennow K, Gauthier S, Valcour V, Miller BL, Rosa-Neto P, Pascoal TA. Neuropsychiatric Symptoms and Microglial Activation in Patients with Alzheimer Disease. JAMA Netw Open 2023; 6:e2345175. [PMID: 38010651 PMCID: PMC10682836 DOI: 10.1001/jamanetworkopen.2023.45175] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/15/2023] [Indexed: 11/29/2023] Open
Abstract
Importance Neuropsychiatric symptoms are commonly encountered and are highly debilitating in patients with Alzheimer disease. Understanding their underpinnings has implications for identifying biomarkers and treatment for these symptoms. Objective To evaluate whether glial markers are associated with neuropsychiatric symptoms in individuals across the Alzheimer disease continuum. Design, Setting, and Participants This cross-sectional study was conducted from January to June 2023, leveraging data from the Translational Biomarkers in Aging and Dementia cohort at McGill University, Canada. Recruitment was based on referrals of individuals from the community or from outpatient clinics. Exclusion criteria included active substance abuse, major surgery, recent head trauma, safety contraindications for positron emission tomography (PET) or magnetic resonance imaging, being currently enrolled in other studies, and having inadequately treated systemic conditions. Main Outcomes and Measures All individuals underwent assessment for neuropsychiatric symptoms (Neuropsychiatry Inventory Questionnaire [NPI-Q]), and imaging for microglial activation ([11C]PBR28 PET), amyloid-β ([18F]AZD4694 PET), and tau tangles ([18F]MK6240 PET). Results Of the 109 participants, 72 (66%) were women and 37 (34%) were men; the median age was 71.8 years (range, 38.0-86.5 years). Overall, 70 had no cognitive impairment and 39 had cognitive impairment (25 mild; 14 Alzheimer disease dementia). Amyloid-β PET positivity was present in 21 cognitively unimpaired individuals (30%) and in 31 cognitively impaired individuals (79%). The NPI-Q severity score was associated with microglial activation in the frontal, temporal, and parietal cortices (β = 7.37; 95% CI, 1.34-13.41; P = .01). A leave-one-out approach revealed that irritability was the NPI-Q domain most closely associated with the presence of brain microglial activation (β = 6.86; 95% CI, 1.77-11.95; P = .008). Furthermore, we found that microglia-associated irritability was associated with study partner burden measured by NPI-Q distress score (β = 5.72; 95% CI, 0.33-11.10; P = .03). Conclusions and Relevance In this cross-sectional study of 109 individuals across the AD continuum, microglial activation was associated with and a potential biomarker of neuropsychiatric symptoms in Alzheimer disease. Moreover, our findings suggest that the combination of amyloid-β- and microglia-targeted therapies could have an impact on relieving these symptoms.
Collapse
Affiliation(s)
- Cristiano Schaffer Aguzzoli
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
- Global Brain Health Institute, University of California, San Francisco
| | - Pâmela C. L. Ferreira
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Guilherme Povala
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - João Pedro Ferrari-Souza
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Bruna Bellaver
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Carolina Soares Katz
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Hussein Zalzale
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Firoza Z. Lussier
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Francieli Rohden
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Sarah Abbas
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Douglas T. Leffa
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Marina Scop Medeiros
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Andréa L. Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Arthur Cassa Macedo
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Vanessa Pallen
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Ann Cohen
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Oscar L. Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Dana L. Tudorascu
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - William E. Klunk
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Victor L. Villemagne
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jean Paul Soucy
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Eduardo R. Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Department of Pharmacology, Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Lucas P. Schilling
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
- Department of Neurology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Thomas K. Karikari
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin–Madison
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Victor Valcour
- Global Brain Health Institute, University of California, San Francisco
- Department of Neurology, University of California, San Francisco
| | - Bruce L. Miller
- Global Brain Health Institute, University of California, San Francisco
- Department of Neurology, University of California, San Francisco
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Tharick A. Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
41
|
Ferrari-Souza JP, Bellaver B, Ferreira PCL, Benedet AL, Povala G, Lussier FZ, Leffa DT, Therriault J, Tissot C, Soares C, Wang YT, Chamoun M, Servaes S, Macedo AC, Vermeiren M, Bezgin G, Kang MS, Stevenson J, Rahmouni N, Pallen V, Poltronetti NM, Cohen A, Lopez OL, Klunk WE, Soucy JP, Gauthier S, Souza DO, Triana-Baltzer G, Saad ZS, Kolb HC, Karikari TK, Villemagne VL, Tudorascu DL, Ashton NJ, Zetterberg H, Blennow K, Zimmer ER, Rosa-Neto P, Pascoal TA. APOEε4 potentiates amyloid β effects on longitudinal tau pathology. NATURE AGING 2023; 3:1210-1218. [PMID: 37749258 PMCID: PMC10592050 DOI: 10.1038/s43587-023-00490-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 08/16/2023] [Indexed: 09/27/2023]
Abstract
The mechanisms by which the apolipoprotein E ε4 (APOEε4) allele influences the pathophysiological progression of Alzheimer's disease (AD) are poorly understood. Here we tested the association of APOEε4 carriership and amyloid-β (Aβ) burden with longitudinal tau pathology. We longitudinally assessed 94 individuals across the aging and AD spectrum who underwent clinical assessments, APOE genotyping, magnetic resonance imaging, positron emission tomography (PET) for Aβ ([18F]AZD4694) and tau ([18F]MK-6240) at baseline, as well as a 2-year follow-up tau-PET scan. We found that APOEε4 carriership potentiates Aβ effects on longitudinal tau accumulation over 2 years. The APOEε4-potentiated Aβ effects on tau-PET burden were mediated by longitudinal plasma phosphorylated tau at threonine 217 (p-tau217+) increase. This longitudinal tau accumulation as measured by PET was accompanied by brain atrophy and clinical decline. Our results suggest that the APOEε4 allele plays a key role in Aβ downstream effects on the aggregation of phosphorylated tau in the living human brain.
Collapse
Affiliation(s)
- João Pedro Ferrari-Souza
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Bruna Bellaver
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Pâmela C L Ferreira
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andréa L Benedet
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Guilherme Povala
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Firoza Z Lussier
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Douglas T Leffa
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Cécile Tissot
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Carolina Soares
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Arthur C Macedo
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Marie Vermeiren
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Gleb Bezgin
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Artificial Intelligence and Computational Neurosciences Laboratory, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Vanessa Pallen
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Nina Margherita Poltronetti
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Ann Cohen
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Oscar L Lopez
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - William E Klunk
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jean-Paul Soucy
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Diogo O Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Ziad S Saad
- Neuroscience Biomarkers, Janssen Research and Development, La Jolla, CA, USA
| | - Hartmuth C Kolb
- Neuroscience Biomarkers, Janssen Research and Development, La Jolla, CA, USA
| | - Thomas K Karikari
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Victor L Villemagne
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dana L Tudorascu
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- UW Department of Medicine, School of Medicine and Public Health, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Graduate Program in Biological Sciences: Pharmacology and Therapeuctis, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Tharick A Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
42
|
Ferreira PCL, Therriault J, Tissot C, Ferrari-Souza JP, Benedet AL, Povala G, Bellaver B, Leffa DT, Brum WS, Lussier FZ, Bezgin G, Servaes S, Vermeiren M, Macedo AC, Cabrera A, Stevenson J, Triana-Baltzer G, Kolb H, Rahmouni N, Klunk WE, Lopez O, Villemagne VL, Cohen A, Tudorascu DL, Zimmer ER, Karikari TK, Ashton NJ, Zetterberg H, Blennow K, Gauthier S, Rosa-Neto P, Pascoal TA. Plasma p-tau231 and p-tau217 inform on tau tangles aggregation in cognitively impaired individuals. Alzheimers Dement 2023; 19:4463-4474. [PMID: 37534889 PMCID: PMC10592380 DOI: 10.1002/alz.13393] [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: 02/24/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 08/04/2023]
Abstract
INTRODUCTION Phosphorylated tau (p-tau) biomarkers have been recently proposed to represent brain amyloid-β (Aβ) pathology. Here, we evaluated the plasma biomarkers' contribution beyond the information provided by demographics (age and sex) to identify Aβ and tau pathologies in individuals segregated as cognitively unimpaired (CU) and impaired (CI). METHODS We assessed 138 CU and 87 CI with available plasma p-tau231, 217+ , and 181, Aβ42/40, GFAP and Aβ- and tau-PET. RESULTS In CU, only plasma p-tau231 and p-tau217+ significantly improved the performance of the demographics in detecting Aβ-PET positivity, while no plasma biomarker provided additional information to identify tau-PET positivity. In CI, p-tau217+ and GFAP significantly contributed to demographics to identify both Aβ-PET and tau-PET positivity, while p-tau231 only provided additional information to identify tau-PET positivity. DISCUSSION Our results support plasma p-tau231 and p-tau217+ as state markers of early Aβ deposition, but in later disease stages they inform on tau tangle accumulation. HIGHLIGHTS It is still unclear how much plasma biomarkers contribute to identification of AD pathology across the AD spectrum beyond the information already provided by demographics (age + sex). Plasma p-tau231 and p-tau217+ contribute to demographic information to identify brain Aβ pathology in preclinical AD. In CI individuals, plasma p-tau231 contributes to age and sex to inform on the accumulation of tau tangles, while p-tau217+ and GFAP inform on both Aβ deposition and tau pathology.
Collapse
Affiliation(s)
- Pamela C. L Ferreira
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Cécile Tissot
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - João Pedro Ferrari-Souza
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Andréa L. Benedet
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
| | - Guilherme Povala
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Bruna Bellaver
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Douglas T. Leffa
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Wagner S. Brum
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
| | - Firoza Z. Lussier
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Marie Vermeiren
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Arthur C. Macedo
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Arlec Cabrera
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Gallen Triana-Baltzer
- Neuroscience Biomarkers, Janssen Research and Development, La Jolla, CA, 92121-1126, USA
| | - Hartmuth Kolb
- Neuroscience Biomarkers, Janssen Research and Development, La Jolla, CA, 92121-1126, USA
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - William E. Klunk
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Oscar Lopez
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Victor L. Villemagne
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Ann Cohen
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Dana L. Tudorascu
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Eduardo R. Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
- Graduate Program in Biological Sciences: Pharmacology and Therapeuctis, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
- Brain Insitute of Rio Grande do Sul, PUCRS, Porto Alegre, 90619-900, Brazil
| | - Thomas K. Karikari
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, 431 41, Sweden
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, WC1N 3BG, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1N 3BG, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, HKG, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
| | - Serge Gauthier
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Pedro Rosa-Neto
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Tharick A. Pascoal
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Neurology, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
43
|
Kim HB, Kim SH, Um YH, Wang SM, Kim REY, Choe YS, Lee J, Kim D, Lim HK, Lee CU, Kang DW. Modulation of associations between education years and cortical volume in Alzheimer's disease vulnerable brain regions by Aβ deposition and APOE ε4 carrier status in cognitively normal older adults. Front Aging Neurosci 2023; 15:1248531. [PMID: 37829142 PMCID: PMC10565031 DOI: 10.3389/fnagi.2023.1248531] [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: 06/28/2023] [Accepted: 09/05/2023] [Indexed: 10/14/2023] Open
Abstract
Background Education years, as a measure of cognitive reserve, have been shown to affect the progression of Alzheimer's disease (AD), both pathologically and clinically. However, inconsistent results have been reported regarding the association between years of education and intermediate structural changes in AD-vulnerable brain regions, particularly when AD risk factors were not considered during the preclinical phase. Objective This study aimed to examine how Aβ deposition and APOE ε4 carrier status moderate the relationship between years of education and cortical volume in AD-vulnerable regions among cognitively normal older adults. Methods A total of 121 participants underwent structural MRI, [18F] flutemetamol PET-CT imaging, and neuropsychological battery assessment. Multiple regression analysis was conducted to examine the interaction between years of education and the effects of potential modifiers on cortical volume. The associations between cortical volume and neuropsychological performance were further explored in subgroups categorized based on AD risk factors. Results The cortical volume of the left lateral occipital cortex and bilateral fusiform gyrus demonstrated a significant differential association with years of education, depending on the presence of Aβ deposition and APOE ε4 carrier status. Furthermore, a significant relationship between the cortical volume of the bilateral fusiform gyrus and AD-nonspecific cognitive function was predominantly observed in individuals without AD risk factors. Conclusion AD risk factors exerted varying influences on the association between years of education and cortical volume during the preclinical phase. Further investigations into the long-term implications of these findings would enhance our understanding of cognitive reserves in the preclinical stages of AD.
Collapse
Affiliation(s)
- Hak-Bin Kim
- Department of Psychiatry, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung-Hwan Kim
- Department of Psychiatry, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoo Hyun Um
- Department of Psychiatry, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Suwon, Republic of Korea
| | - Sheng-Min Wang
- Department of Psychiatry, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | | | - Yeong Sim Choe
- Research Institute, NEUROPHET Inc., Seoul, Republic of Korea
| | - Jiyeon Lee
- Research Institute, NEUROPHET Inc., Seoul, Republic of Korea
| | - Donghyeon Kim
- Research Institute, NEUROPHET Inc., Seoul, Republic of Korea
| | - Hyun Kook Lim
- Department of Psychiatry, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Research Institute, NEUROPHET Inc., Seoul, Republic of Korea
| | - Chang Uk Lee
- Department of Psychiatry, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dong Woo Kang
- Department of Psychiatry, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
44
|
Kunach P, Vaquer-Alicea J, Smith MS, Hopewell R, Monistrol J, 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. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.22.558671. [PMID: 37790438 PMCID: PMC10542181 DOI: 10.1101/2023.09.22.558671] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Positron Emission Tomography (PET) ligands have advanced Alzheimer's disease (AD) diagnosis and treatment. Using autoradiography and cryo-EM, we identified AD brain tissue with elevated tau burden, purified filaments, and determined 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 K353, 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, Quebec, Canada
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, Dallas, TX, United States
| | - Jaime Vaquer-Alicea
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, Dallas, TX, United States
| | - Matthew S. Smith
- Department of Pharmaceutical Chemistry, UCSF, San Francisco, CA, United States
- Program of Biophysics, UCSF, San Francisco, CA, United States
| | | | - Jim Monistrol
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, Dallas, TX, United States
| | - Luc Moquin
- Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Joseph Therriault
- Department of Neurology, McGill University, Montreal, Quebec, Canada
| | - Cecile Tissot
- Department of Neurology, McGill University, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- Department of Neurology, McGill University, Montreal, Quebec, Canada
| | | | | | - Marie-Christine Guiot
- Department of Neurology, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Brian K. Shoichet
- Department of Pharmaceutical Chemistry, UCSF, San Francisco, CA, United States
| | - Pedro Rosa-Neto
- Department of Neurology, McGill University, Montreal, Quebec, Canada
| | - Marc I. Diamond
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, Dallas, TX, United States
| | - Sarah H. Shahmoradian
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, Dallas, TX, United States
| |
Collapse
|
45
|
Li J, Kumar A, Långström B, Nordberg A, Ågren H. Insight into the Binding of First- and Second-Generation PET Tracers to 4R and 3R/4R Tau Protofibrils. ACS Chem Neurosci 2023; 14:3528-3539. [PMID: 37639522 PMCID: PMC10515481 DOI: 10.1021/acschemneuro.3c00437] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023] Open
Abstract
Primary supranuclear palsy (PSP) is a rare neurodegenerative disease that perturbs body movement, eye movement, and walking balance. Similar to Alzheimer's disease (AD), the abnormal aggregation of tau fibrils in the central neuronal and glial cells is a major hallmark of PSP disease. In this study, we use multiple approaches, including docking, molecular dynamics, and metadynamics simulations, to investigate the binding mechanism of 10 first- and second-generations of PET tracers for PSP tau and compare their binding in cortical basal degeneration (CBD) and AD tauopathies. Structure-activity relationships, binding preferences, the nature of ligand binding in terms of basic intermolecular interactions, the role of polar/charged residues, induced-fit mechanisms, grove closures, and folding patterns for the binding of these tracers in PSP, CBD, and AD tau fibrils are evaluated and discussed in detail in order to build a holistic picture of what is essential for the binding and also to rank the potency of the different tracers. For example, we found that the same tracer shows different binding preferences for the surface sites of tau fibrils that are intrinsically distinct in the folding patterns. Results from the metadynamics simulations predict that PMPBB3 and PBB3 exhibit the strongest binding free energies onto the Q276[I277]I278, Q351[S352]K353, and N368[K369]K370 sites of PSP than the other explored tracers, indicating a solid preference for vdW and cation-π interactions. Our results also reproduced known preferences of tracers, namely, that MK6240 binds better to AD tau than CBD tau and PSP tau and that CBD2115, PI2620, and PMPBB3 are 4R tau binders. These findings fill in the well-sought-after knowledge gap in terms of these tracers' potential binding mechanisms and will be important for the design of highly selective novel PET tracers for tauopathies.
Collapse
Affiliation(s)
- Junhao Li
- Department
of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden
| | - Amit Kumar
- Department
of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Neo, 141 84 Stockholm, Sweden
| | - Bengt Långström
- Department
of Chemistry - BMC, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden
| | - Agneta Nordberg
- Department
of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Neo, 141 84 Stockholm, Sweden
- Theme
Inflammation and Aging, Karolinska University
Hospital, S-141 86 Stockholm, Sweden
| | - Hans Ågren
- Department
of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden
- College
of Chemistry and Chemical Engineering, Henan
University, Kaifeng, Henan 475004, P. R. China
| |
Collapse
|
46
|
Harada R, Lerdsirisuk P, Shimizu Y, Yokoyama Y, Du Y, Kudo K, Ezura M, Ishikawa Y, Iwata R, Shidahara M, Ishiki A, Kikuchi A, Hatano Y, Ishihara T, Onodera O, Iwasaki Y, Yoshida M, Taki Y, Arai H, Kudo Y, Yanai K, Furumoto S, Okamura N. Preclinical Characterization of the Tau PET Tracer [ 18F]SNFT-1: Comparison of Tau PET Tracers. J Nucl Med 2023; 64:1495-1501. [PMID: 37321821 DOI: 10.2967/jnumed.123.265593] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/03/2023] [Indexed: 06/17/2023] Open
Abstract
Tau PET tracers are expected to be sufficiently sensitive to track the progression of age-related tau pathology in the medial temporal cortex. The tau PET tracer N-(4-[18F]fluoro-5-methylpyridin-2-yl)-7-aminoimidazo[1,2-a]pyridine ([18F]SNFT-1) has been successfully developed by optimizing imidazo[1,2-a]pyridine derivatives. We characterized the binding properties of [18F]SNFT-1 using a head-to-head comparison with other reported 18F-labeled tau tracers. Methods: The binding affinity of SNFT-1 to tau, amyloid, and monoamine oxidase A and B was compared with that of the second-generation tau tracers MK-6240, PM-PBB3, PI-2620, RO6958948, JNJ-64326067, and flortaucipir. In vitro binding properties of 18F-labeled tau tracers were evaluated through the autoradiography of frozen human brain tissues from patients with diverse neurodegenerative disease spectra. Pharmacokinetics, metabolism, and radiation dosimetry were assessed in normal mice after intravenous administration of [18F]SNFT-1. Results: In vitro binding assays demonstrated that [18F]SNFT-1 possesses high selectivity and high affinity for tau aggregates in Alzheimer disease (AD) brains. Autoradiographic analysis of tau deposits in medial temporal brain sections from patients with AD showed a higher signal-to-background ratio for [18F]SNFT-1 than for the other tau PET tracers and no significant binding with non-AD tau, α-synuclein, transactiviation response DNA-binding protein-43, and transmembrane protein 106B aggregates in human brain sections. Furthermore, [18F]SNFT-1 did not bind significantly to various receptors, ion channels, or transporters. [18F]SNFT-1 showed a high initial brain uptake and rapid washout from the brains of normal mice without radiolabeled metabolites. Conclusion: These preclinical data suggest that [18F]SNFT-1 is a promising and selective tau radiotracer candidate that allows the quantitative monitoring of age-related accumulation of tau aggregates in the human brain.
Collapse
Affiliation(s)
- Ryuichi Harada
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan;
- Division of Brain Science, Department of Aging Research and Geriatric Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | | | - Yuki Shimizu
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Yuka Yokoyama
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Yiqing Du
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kaede Kudo
- Division of Brain Science, Department of Aging Research and Geriatric Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Michinori Ezura
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoichi Ishikawa
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Ren Iwata
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Miho Shidahara
- Department of Quantum Science and Energy Engineering, Tohoku University, Sendai, Japan
| | - Aiko Ishiki
- Division of Brain Science, Department of Aging Research and Geriatric Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
- Division of Community Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Akio Kikuchi
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuya Hatano
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Tomohiko Ishihara
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yasushi Iwasaki
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Japan; and
| | - Mari Yoshida
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Japan; and
| | - Yasuyuki Taki
- Division of Brain Science, Department of Aging Research and Geriatric Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Hiroyuki Arai
- Division of Brain Science, Department of Aging Research and Geriatric Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Yukitsuka Kudo
- Division of Brain Science, Department of Aging Research and Geriatric Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Kazuhiko Yanai
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Shozo Furumoto
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Nobuyuki Okamura
- Division of Brain Science, Department of Aging Research and Geriatric Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| |
Collapse
|
47
|
Bellaver B, Puig-Pijoan A, Ferrari-Souza JP, Leffa DT, Lussier FZ, Ferreira PCL, Tissot C, Povala G, Therriault J, Benedet AL, Ashton NJ, Servaes S, Chamoun M, Stevenson J, Rahmouni N, Vermeiren M, Macedo AC, Fernández-Lebrero A, García-Escobar G, Navalpotro-Gómez I, Lopez O, Tudorascu DL, Cohen A, Villemagne VL, Klunk WE, Gauthier S, Zimmer ER, Karikari TK, Blennow K, Zetterberg H, Suárez-Calvet M, Rosa-Neto P, Pascoal TA. Blood-brain barrier integrity impacts the use of plasma amyloid-β as a proxy of brain amyloid-β pathology. Alzheimers Dement 2023; 19:3815-3825. [PMID: 36919582 PMCID: PMC10502181 DOI: 10.1002/alz.13014] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/08/2022] [Accepted: 01/25/2023] [Indexed: 03/16/2023]
Abstract
INTRODUCTION Amyloid-β (Aβ) and tau can be quantified in blood. However, biological factors can influence the levels of brain-derived proteins in the blood. The blood-brain barrier (BBB) regulates protein transport between cerebrospinal fluid (CSF) and blood. BBB altered permeability might affect the relationship between brain and blood biomarkers. METHODS We assessed 224 participants in research (TRIAD, n = 96) and clinical (BIODEGMAR, n = 128) cohorts with plasma and CSF/positron emission tomography Aβ, p-tau, and albumin measures. RESULTS Plasma Aβ42/40 better identified CSF Aβ42/40 and Aβ-PET positivity in individuals with high BBB permeability. An interaction between plasma Aβ42/40 and BBB permeability on CSF Aβ42/40 was observed. Voxel-wise models estimated that the association of positron emission tomography (PET), with plasma Aβ was most affected by BBB permeability in AD-related brain regions. BBB permeability did not significantly impact the relationship between brain and plasma p-tau levels. DISCUSSION These findings suggest that BBB integrity may influence the performance of plasma Aβ, but not p-tau, biomarkers in research and clinical settings. HIGHLIGHTS BBB permeability affects the association between brain and plasma Aβ levels. BBB integrity does not affect the association between brain and plasma p-tau levels. Plasma Aβ was most affected by BBB permeability in AD-related brain regions. BBB permeability increases with age but not according to cognitive status.
Collapse
Affiliation(s)
- Bruna Bellaver
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Albert Puig-Pijoan
- Cognitive Decline and Movement Disorders Unit, Neurology Department, Hospital del Mar, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - João Pedro Ferrari-Souza
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Douglas T Leffa
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Firoza Z Lussier
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Pamela C L Ferreira
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Guilherme Povala
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Andréa L Benedet
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Marie Vermeiren
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Arthur C Macedo
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Aida Fernández-Lebrero
- Cognitive Decline and Movement Disorders Unit, Neurology Department, Hospital del Mar, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
| | | | - Irene Navalpotro-Gómez
- Cognitive Decline and Movement Disorders Unit, Neurology Department, Hospital del Mar, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
| | - Oscar Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Dana L Tudorascu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ann Cohen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Victor L Villemagne
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - William E Klunk
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Thomas K Karikari
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Marc Suárez-Calvet
- Cognitive Decline and Movement Disorders Unit, Neurology Department, Hospital del Mar, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
48
|
Wagatsuma K, Miwa K, Akamatsu G, Yamao T, Kamitaka Y, Sakurai M, Fujita N, Hanaoka K, Matsuda H, Ishii K. Toward standardization of tau PET imaging corresponding to various tau PET tracers: a multicenter phantom study. Ann Nucl Med 2023; 37:494-503. [PMID: 37243882 DOI: 10.1007/s12149-023-01847-8] [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: 03/20/2023] [Accepted: 05/17/2023] [Indexed: 05/29/2023]
Abstract
OBJECTIVE Tau positron emission tomography (PET) imaging is a recently developed non-invasive tool that can detect the density and extension of tau neurofibrillary tangles. Tau PET tracers have been validated to harmonize and accelerate their development and implementation in clinical practice. Whereas standard protocols including injected dose, uptake time, and duration have been determined for tau PET tracers, reconstruction parameters have not been standardized. The present study conducted phantom experiments based on tau pathology to standardize quantitative tau PET imaging parameters and optimize reconstruction conditions of PET scanners at four Japanese sites according to the results of phantom experiments. METHODS The activity of 4.0 and 2.0 kBq/mL for Hoffman 3D brain and cylindrical phantoms, respectively, was estimated from published studies of brain activity using [18F]flortaucipir, [18F]THK5351, and [18F]MK6240. We developed an original tau-specific volume of interest template for the brain based on pathophysiological tau distribution in the brain defined as Braak stages. We acquired brain and cylindrical phantom images using four PET scanners. Iteration numbers were determined as contrast and recover coefficients (RCs) in gray (GM) and white (WM) matter, and the magnitude of the Gaussian filter was determined from image noise. RESULTS Contrast and RC converged at ≥ 4 iterations, the error rates of RC for GM and WM were < 15% and 1%, respectively, and noise was < 10% in Gaussian filters of 2-4 mm in images acquired using the four scanners. Optimizing the reconstruction conditions for phantom tau PET images acquired by each scanner improved contrast and image noise. CONCLUSIONS The phantom activity was comprehensive for first- and second-generation tau PET tracers. The mid-range activity that we determined could be applied to later tau PET tracers. We propose an analytical tau-specific VOI template based on tau pathophysiological changes in patients with AD to standardize tau PET imaging. Phantom images reconstructed under the optimized conditions for tau PET imaging achieved excellent image quality and quantitative accuracy.
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 of Gerontology, 35-2, Sakae-Cho, Itabashi-Ku, Tokyo, 173-0015, Japan.
| | - Kenta Miwa
- Department of Radiological Sciences, School of Health Sciences, Fukushima Medical University, Fukushima City, Fukushima, 960-1295, 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
| | - Tensho Yamao
- Department of Radiological Sciences, School of Health Sciences, Fukushima Medical University, Fukushima City, Fukushima, 960-1295, Japan
| | - Yuto Kamitaka
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2, Sakae-Cho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Minoru Sakurai
- Clinical Imaging Center for Healthcare, Nippon Medical School, 1-12-15, Sendagi, Bunkyo-Ku, Tokyo, 113-0022, Japan
| | - Naotoshi Fujita
- Department of Radiological Technology, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8560, Japan
| | - Kohei Hanaoka
- Division of Positron Emission Tomography, Institute of Advanced Clinical Medicine, Kindai University, 377-2 Onohigashi, Osakasayama, Osaka, 589-8511, Japan
| | - Hiroshi Matsuda
- Department of Biofunctional Imaging, Fukushima Medical University, 1 Hikarigaoka, Fukushima City, Fukushima, 960-1295, Japan
- Drug Discovery and Cyclotron Research Center, Southern Tohoku Research Institute for Neuroscience, 7-115, Yatsuyamada, Koriyama, 963-8052, Japan
| | - Kenji Ishii
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2, Sakae-Cho, Itabashi-Ku, Tokyo, 173-0015, Japan
| |
Collapse
|
49
|
Matsuda H, Yamao T. Tau positron emission tomography in patients with cognitive impairment and suspected Alzheimer's disease. Fukushima J Med Sci 2023; 69:85-93. [PMID: 37302841 PMCID: PMC10480511 DOI: 10.5387/fms.2023-08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/10/2023] [Indexed: 06/13/2023] Open
Abstract
Alzheimer's disease (AD) is diagnosed by the presence of both amyloid β and tau proteins. Recent advances in molecular PET imaging have made it possible to assess the accumulation of these proteins in the living brain. PET ligands have been developed that bind to 3R/4R tau in AD, but not to 3R tau or 4R tau alone. Of the first-generation PET ligands, 18F-flortaucipir has recently been approved by the Food and Drug Administration. Several second-generation PET probes with less off-target binding have been developed and are being applied clinically. Visual interpretation of tau PET should be based on neuropathological neurofibrillary tangle staging instead of a simple positive or negative classification. Four visual read classifications have been proposed: "no uptake," "medial temporal lobe (MTL) only," "MTL AND," and "outside MTL." As an adjunct to visual interpretation, quantitative analysis has been proposed using MRI-based native space FreeSurfer parcellations. The standardized uptake value ratio of the target area is measured using the cerebellar gray matter as a reference region. In the near future, the Centiloid scale of tau PET is expected to be used as a harmonized value for standardizing each analytical method or PET ligand used, similar to amyloid PET.
Collapse
Affiliation(s)
- Hiroshi Matsuda
- Department of Biofunctional Imaging, Fukushima Medical University
| | - Tensho Yamao
- Department of Radiological Sciences, School of Health Sciences, Fukushima Medical University
| |
Collapse
|
50
|
Sanchez-Rodriguez LM, Bezgin G, Carbonell F, Therriault J, Fernandez-Arias J, Servaes S, Rahmouni N, Tissot C, Stevenson J, Karikari TK, Ashton NJ, Benedet AL, Zetterberg H, Blennow K, Triana-Baltzer G, Kolb HC, Rosa-Neto P, Iturria-Medina Y. Revealing the combined roles of Aβ and tau in Alzheimer's disease via a pathophysiological activity decoder. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.21.529377. [PMID: 37502947 PMCID: PMC10370127 DOI: 10.1101/2023.02.21.529377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Neuronal dysfunction and cognitive deterioration in Alzheimer's disease (AD) are likely caused by multiple pathophysiological factors. However, evidence in humans remains scarce, necessitating improved non-invasive techniques and integrative mechanistic models. Here, we introduce personalized brain activity models incorporating functional MRI, amyloid-β (Aβ) and tau-PET from AD-related participants ( N = 132 ) . Within the model assumptions, electrophysiological activity is mediated by toxic protein deposition. Our integrative subject-specific approach uncovers key patho-mechanistic interactions, including synergistic Aβ and tau effects on cognitive impairment and neuronal excitability increases with disease progression. The data-derived neuronal excitability values strongly predict clinically relevant AD plasma biomarker concentrations (p-tau217, p-tau231, p-tau181, GFAP). Furthermore, our results reproduce hallmark AD electrophysiological alterations (theta band activity enhancement and alpha reductions) which occur with Aβ-positivity and after limbic tau involvement. Microglial activation influences on neuronal activity are less definitive, potentially due to neuroimaging limitations in mapping neuroprotective vs detrimental phenotypes. Mechanistic brain activity models can further clarify intricate neurodegenerative processes and accelerate preventive/treatment interventions.
Collapse
Affiliation(s)
- Lazaro M. Sanchez-Rodriguez
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada
| | - Gleb Bezgin
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, Canada
| | | | - Joseph Therriault
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, Canada
| | - Jaime Fernandez-Arias
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, Canada
| | - Stijn Servaes
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, Canada
| | - Nesrine Rahmouni
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, Canada
| | - Cecile Tissot
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, Canada
| | - Jenna Stevenson
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, Canada
| | - Thomas K. Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience Maurice Wohl Institute Clinical Neuroscience Institute London UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation London UK
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Andréa L. Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal
| | | | - Hartmuth C. Kolb
- Neuroscience Biomarkers, Janssen Research & Development, La Jolla, California, USA
| | - Pedro Rosa-Neto
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, Canada
| | - Yasser Iturria-Medina
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada
| |
Collapse
|