1
|
Mao C, Mo Y, Jiang J, Fang S, Hu Z, Ke Z, Zhao H, Xu Y. Association between high plasma p-tau181 level and gait changes in patients with mild cognitive impairment. Sci Rep 2025; 15:14679. [PMID: 40287471 PMCID: PMC12033327 DOI: 10.1038/s41598-025-94472-6] [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: 08/06/2024] [Accepted: 03/13/2025] [Indexed: 04/29/2025] Open
Abstract
Previous studies on gait changes in mild cognitive impairment (MCI) are inconsistent. Alzheimer's disease (AD) plasma biomarkers, amyloid beta (Aβ) and phosphorylated-tau (p-tau), are relevant to gait disorders. This study explores gait changes in MCI and the relationship between gait performance and AD plasma biomarkers. 231 participants were recruited and stratified based on p-tau181 levels into: low p-tau181 with normal cognition (lT-NC), low p-tau181 with MCI (lT-MCI), and high p-tau181 with MCI (hT-MCI). The same cohort was subsequently stratified by Aβ42/Aβ40 levels into: high Aβ42/Aβ40 with normal cognition (hA-NC), high Aβ42/Aβ40 with MCI (hA-MCI), and low Aβ42/Aβ40 with MCI (lA-MCI). Demographic, cognitive and gait data were compared across groups. The hT-MCI and lA-MCI groups were older than the other groups. Significant differences in stride length were found between lT-NC and hT-MCI, lT-MCI and hT-MCI, but not between lT-NC and lT-MCI. Neuropsychological assessments revealed poorer performance in hT-MCI and lT-MCI groups relative to lT-NC, while global cognitive function was comparable between hT-MCI and lT-MCI groups. No such associations were observed between stride length and Aβ42/Aβ40 levels. Decreased stride length, which is generally considered to be indicative of poorer gait, was significantly associated with elevated p-tau181 levels and independent of global cognitive status. These findings highlight the potential of p-tau181 as a biomarker for tau-related motor dysfunction in MCI.
Collapse
Affiliation(s)
- Chenglu Mao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China.
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210000, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210000, China.
- Nanjing Neurology Clinical Medical Center, Nanjing, 210000, China.
| | - Yuting Mo
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210000, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210000, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210000, China
- Nanjing Neurology Clinical Medical Center, Nanjing, 210000, China
| | - Jialiu Jiang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210000, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210000, China
- Nanjing Neurology Clinical Medical Center, Nanjing, 210000, China
| | - Shuang Fang
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210000, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210000, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210000, China
- Nanjing Neurology Clinical Medical Center, Nanjing, 210000, China
| | - Zheqi Hu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210000, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210000, China
- Nanjing Neurology Clinical Medical Center, Nanjing, 210000, China
| | - Zhihong Ke
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210000, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210000, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210000, China
- Nanjing Neurology Clinical Medical Center, Nanjing, 210000, China
| | - Hui Zhao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China.
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210000, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210000, China.
- Nanjing Neurology Clinical Medical Center, Nanjing, 210000, China.
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China.
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210000, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210000, China.
- Nanjing Neurology Clinical Medical Center, Nanjing, 210000, China.
| |
Collapse
|
2
|
Furuya Y, Sato K, Niimi Y, Ihara R, Suzuki K, Iwata A, Iwatsubo T. Cognitive function instrument-based anosognosia to predict amyloid status. J Alzheimers Dis 2025; 104:995-1000. [PMID: 39924909 DOI: 10.1177/13872877251317508] [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: 02/11/2025]
Abstract
Anosognosia, a lack of self-awareness regarding cognitive dysfunction, often accompanies the progression of Alzheimer's disease (AD) pathology. This study explored the relationship between AD pathology and anosognosia measured by discrepancies in Cognitive Function Instrument (CFI) scores, as rated by participants and their study partners (SP). Using mixed-effects models on non-demented participant data, the results revealed that lower self-reported CFI score compared to SP ratings was significantly associated with positive amyloid PET results (odds ratio 1.081 per-1 decrease in ΔCFI). Our findings suggest that CFI-based anosognosia could serve as a potential predictor of positive amyloid PET status.
Collapse
Affiliation(s)
- Yuichiro Furuya
- Division of Neurology, Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Kenichiro Sato
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Unit for Early and Exploratory Development, The University of Tokyo Hospital, Tokyo, Japan
| | - Yoshiki Niimi
- Unit for Early and Exploratory Development, The University of Tokyo Hospital, Tokyo, Japan
- Department of Healthcare Economics and Health Policy, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryoko Ihara
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Kazushi Suzuki
- Division of Neurology, Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Atsushi Iwata
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Unit for Early and Exploratory Development, The University of Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
3
|
Abuwarda H, Trainer A, Horien C, Shen X, Ju S, Constable RT, Fredericks C. Whole-brain functional connectivity predicts regional tau PET in preclinical Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.02.587791. [PMID: 38617320 PMCID: PMC11014551 DOI: 10.1101/2024.04.02.587791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Preclinical Alzheimer's disease (AD), characterized by the abnormal accumulation of amyloid prior to cognitive symptoms, presents a critical opportunity for early intervention. Past work has described functional connectivity changes in preclinical disease, yet the interplay between AD pathology and the functional connectome during this window remains unexplored. We applied connectome-based predictive modeling to investigate the ability of resting-state whole-brain functional connectivity to predict tau (18F-flortaucipir) and amyloid (18F-florbetapir) PET binding in a preclinical AD cohort (A4, n =342, age 65-85). Separate predictive models were developed for each of 14 regions, and model performance was assessed using a Spearman's correlation between predicted and observed PET binding standard uptake value ratios. We assessed the validity of significant models by applying them to an external dataset, and visualized the underlying connectivity that was positively and negatively correlated to posterior cingulate tau binding, the most successful model. We found that whole brain functional connectivity predicts regional tau PET, outperforming amyloid PET models. The best performing tau models were for regions affected in Braak stage IV-V regions (posterior cingulate, precuneus, lateral occipital cortex, middle temporal, inferior temporal, and Bank STS), while models for regions of earlier tau pathology (entorhinal, parahippocampal, fusiform, and amygdala) performed poorly. Importantly, tau models generalized to a symptomatic AD cohort (ADNI; amyloid positive, n = 211, age 55-90), in tau-elevated but not tau-negative individuals. For the posterior cingulate A4 tau model, the most successful model, the predictive edges positively correlated with posterior cingulate tau predominantly came from nodes within temporal, limbic, and cerebellar regions. The most predictive edges negatively associated to tau were from nodes of heteromodal association areas, particularly within the prefrontal and parietal cortices. These findings reveal that whole-brain functional connectivity predicts tau PET in preclinical AD and generalizes to a clinical dataset specifically in individuals with abnormal tau PET, highlighting the relevance of the functional connectome for the early detection and monitoring of AD pathology.
Collapse
|
4
|
Kuhn E, Klinger HM, Amariglio RE, Wagner M, Jessen F, Düzel E, Heneka MT, Chételat G, Rentz DM, Sperling RA, Ebenau JL, Butterbrod E, Van Der Flier WM, Sikkes SAM, Teunnissen CE, Van Harten AC, Van De Giessen EM, Rami L, Tort A, Sánchez Benavides G, Gifford KA, Van Hulle C, Buckley RF. SCD-plus features and AD biomarkers in cognitively unimpaired samples: A meta-analytic approach for nine cohort studies. Alzheimers Dement 2025. [PMID: 39985404 DOI: 10.1002/alz.14307] [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: 05/29/2024] [Accepted: 09/10/2024] [Indexed: 02/24/2025]
Abstract
INTRODUCTION Specific features of subjective cognitive decline (SCD-plus) have been proposed to indicate an increased risk of preclinical Alzheimer's disease (AD). However, few studies have examined how these features relate to AD biomarkers in cognitively unimpaired (CU) older adults. METHODS Meta-analyses were performed using cross-sectional data from nine cohorts (n = 7219, mean age (SD): 71.17 (5.9), 56.5% female) to determine associations of SCD-plus features with positron emission tomography (PET)- or cerebrospinal fluid (CSF)-derived amyloid beta (Aβ) and tau biomarkers. RESULTS Participants with preclinical AD (community-based only) were more likely to fulfill SCD-plus features. The presence of self-reported memory decline, associated concern/worry, and a higher number of fulfilled features were all associated with high Aβ levels. Only the latter was associated with abnormal tau. DISCUSSION Simultaneous endorsement of multiple SCD-plus features is a robust indicator of abnormal AD biomarkers in CU older adults, whereas isolated SCD features seem only sensitive to elevated Aβ, supporting their value as early behavioral markers of preclinical AD. HIGHLIGHTS About two-tenths of our sample had abnormal amyloid beta (Aβ) levels with evidence of subjective cognitive decline (SCD). Preclinical AD subsamples (community-based) had a higher percentage of participants meeting SCD-plus features. Self-reported memory decline and concern/worry were the sole features associated with high Aβ, but not tau, burden. A higher number of fulfilled SCD-plus features are linked to high Aβ and tau burden. Use of multiple SCD-plus features may help identify early stages of biological AD.
Collapse
Grants
- BN012 Deutsches Zentrum für Neurodegenerative Erkrankungen
- W81XWH-12-2-0012 U.S. Department of Defense
- DP2AG082342 NIA NIH HHS
- University Caen Normandy
- INSERM
- Fondation Philippe Chatrier
- ZT-I-PF-5-163 Helmholtz Artificial Intelligence Cooperation Unit
- R00-AG061238 National Institutes of Health/National Institute on Aging (NIH/NIA)
- DP2AG082342 National Institutes of Health/National Institute on Aging (NIH/NIA)
- R01-AG079142 National Institutes of Health/National Institute on Aging (NIH/NIA)
- K23-AG045966 National Institutes of Health/National Institute on Aging (NIH/NIA)
- R01-AG062826 National Institutes of Health/National Institute on Aging (NIH/NIA)
- U01AG024904 National Institutes of Health/National Institute on Aging (NIH/NIA)
- R01-AG063689 National Institutes of Health/National Institute on Aging (NIH/NIA)
- U19AG010483 National Institutes of Health/National Institute on Aging (NIH/NIA)
- U24AG057437 National Institutes of Health/National Institute on Aging (NIH/NIA)
- P01AG036694 National Institutes of Health/National Institute on Aging (NIH/NIA)
- R01-AG027161 National Institutes of Health/National Institute on Aging (NIH/NIA)
- UL1TR000427 NIH-NCATS
- CP23/00039 Instituto de Salud Carlos III
- European Union, FSE+
- NIBIB NIH HHS
- AbbVie
- IIRG-08-88733 Alzheimer's Association
- Alzheimer's Drug Discovery Foundation
- Araclon Biotech
- BioClinica, Inc.
- Biogen
- Bristol-Myers Squibb Company
- CereSpir, Inc.
- Cogstate
- Eisai Inc.
- Elan Pharmaceuticals, Inc.
- Eli Lilly and Company
- EuroImmun
- F. Hoffmann-La Roche Ltd and Genentech, Inc.
- Fujirebio
- GE Healthcare
- IXICO Ltd.
- Janssen Alzheimer Immunotherapy Research & Development, LLC
- Johnson & Johnson Pharmaceutical Research & Development LLC
- Lumosity
- Lundbeck
- Merck & Co., Inc.
- Meso Scale Diagnostics, LLC
- NeuroRx Research
- Neurotrack Technologies
- Novartis Pharmaceuticals Corporation
- Pfizer Inc.
- Piramal Imaging
- Servier
- Takeda Pharmaceutical Company
- Transition Therapeutics
- CIHR
- Foundation for the National Institutes of Health
- Northern California Institute for Research and Education
- Alzheimer's Therapeutic Research Institute, University of Southern California
- Laboratory for Neuro Imaging, University of Southern California
- Austin Health
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)
- Edith Cowan University
- Florey Institute, The University of Melbourne
- National Ageing Research Institute
- GHR Foundation
- Davis Alzheimer Prevention Program
- Brigham and Women's Hospital
- Albert Einstein College of Medicine
- Foundation for Neurologic Disease
- 2011-A01493-38 PHRCN
- 2012-12-006-0347 PHRCN
- Agence Nationale de la Recherche (ANR LONGVIE 2007)
- Fondation Plan Alzheimer (Alzheimer Plan 2008-2012)
- Association France Alzheimer et maladies apparentées (AAP 2013)
- Région Basse Normandie
- Dioraphte and the Noaber Foundation
- AVID
- Pasman Chair
Collapse
Affiliation(s)
- Elizabeth Kuhn
- German Center for Neurodegenerative Diseases (DZNE) Bonn, Bonn, Germany
- Department of Cognitive Disorders and Old Age Psychiatry, University Hospital Bonn, Bonn, Germany
| | - Hannah M Klinger
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Rebecca E Amariglio
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Center for Alzheimer Research and Treatment (CART), Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE) Bonn, Bonn, Germany
- Department of Cognitive Disorders and Old Age Psychiatry, University Hospital Bonn, Bonn, Germany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE) Bonn, Bonn, Germany
- Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Universitätsplatz 2, Magdeburg, Germany
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Esch-sur-Alzette, Luxembourg
| | - Gael Chételat
- Normandie Univ, UNICAEN, INSERM, U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Neuropresage Team, Cyceron, Caen cedex, France
| | - Dorene M Rentz
- Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Center for Alzheimer Research and Treatment (CART), Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Jarith L Ebenau
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Elke Butterbrod
- Department of Clinical, Neuro and Developmental Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Neurosurgery, Elisabeth-Tweesteden Hospital, Tilburg, 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
- Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Sietske A M Sikkes
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Department of Clinical, Neuro and Developmental Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Charlotte E Teunnissen
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
| | - Argonde C Van Harten
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Elsmarieke M Van De Giessen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
| | - Lorena Rami
- Hospital Clinic. Fundació Clinic, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Adria Tort
- Hospital Clinic. Fundació Clinic, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | | | - Katherine A Gifford
- Vanderbilt Memory and Alzheimer's Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Carol Van Hulle
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Rachel F Buckley
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Center for Alzheimer Research and Treatment (CART), Brigham & Women's Hospital, Boston, Massachusetts, USA
- Melbourne School of Psychological Sciences, University of Melbourne, Melbourne, Australia
| |
Collapse
|
5
|
Davis MR, Robinson E, Koronyo Y, Salobrar-Garcia E, Rentsendorj A, Gaire BP, Mirzaei N, Kayed R, Sadun AA, Ljubimov AV, Schneider LS, Hawes D, Black KL, Fuchs DT, Koronyo-Hamaoui M. Retinal ganglion cell vulnerability to pathogenic tau in Alzheimer's disease. Acta Neuropathol Commun 2025; 13:31. [PMID: 39955563 PMCID: PMC11829413 DOI: 10.1186/s40478-025-01935-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 01/25/2025] [Indexed: 02/17/2025] Open
Abstract
Pathological tau isoforms, including hyperphosphorylated tau at serine 396 (pS396-tau) and tau oligomers (Oligo-tau), are elevated in the retinas of patients with mild cognitive impairment (MCI) due to Alzheimer's disease (AD) and AD dementia. These patients exhibit significant retinal ganglion cell (RGC) loss, however the presence of tau isoforms in RGCs and their impact on RGC integrity, particularly in early AD, have not been studied. Here, we analyzed retinal superior temporal cross-sections from 25 MCI or AD patients and 16 age- and sex-matched cognitively normal controls. Using the RGC marker ribonucleic acid binding protein with multiple splicing (RBPMS) and Nissl staining, we found a 46-56% reduction in RBPMS+ RGCs and Nissl+ neurons in the ganglion cell layer (GCL) of MCI and AD retinas (P < 0.05-0.001). RGC loss was accompanied by soma hypertrophy (10-50% enlargement, P < 0.05-0.0001), nuclear displacement, apoptosis (30-50% increase, P < 0.05-0.01), and prominent expression of granulovacuolar degeneration (GVD) bodies and GVD-necroptotic markers. Both pS396-tau and Oligo-tau were identified in RGCs, including in hypertrophic cells. PS396-tau+ and Oligo-tau+ RGC counts were significantly increased by 2.1-3.5-fold in MCI and AD retinas versus control retinas (P < 0.05-0.0001). Tauopathy-laden RGCs strongly inter-correlated (rP=0.85, P < 0.0001) and retinal tauopathy associated with RGC reduction (rP=-0.40-(-0.64), P < 0.05-0.01). Their abundance correlated with brain pathology and cognitive deficits, with higher tauopathy-laden RGCs in patients with Braak stages (V-VI), clinical dementia ratings (CDR = 3), and mini-mental state examination (MMSE ≤ 26) scores. PS396-tau+ RGCs in the central and mid-periphery showed the closest associations with disease status, while Oligo-tau+ RGCs in the mid-periphery exhibited the strongest correlations with brain pathology (NFTs, Braak stages, ABC scores; rS=0.78-0.81, P < 0.001-0.0001) and cognitive decline (MMSE; rS=-0.79, P = 0.0019). Overall, these findings identify a link between pathogenic tau in RGCs and RGC degeneration in AD, involving apoptotic and GVD-necroptotic cell death pathways. Future research should validate these results in larger and more diverse cohorts and develop RGC tauopathy as a potential noninvasive biomarker for early detection and monitoring of AD progression.
Collapse
Affiliation(s)
- Miyah R Davis
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
| | - Edward Robinson
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
| | - Elena Salobrar-Garcia
- Institute of Ophthalmologic Research Ramón Castroviejo, Complutense University of Madrid, Madrid, 28040, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, 28040, Spain
- Health Research Institute, Clinico San Carlos Hospital (IdISSC), Madrid, 28040, Spain
| | - Altan Rentsendorj
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
| | - Bhakta P Gaire
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
| | - Nazanin Mirzaei
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Alfredo A Sadun
- Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Doheny Eye Institute, Los Angeles, CA, USA
| | - Alexander V Ljubimov
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
- Eye Program, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars- Sinai Medical Center, Los Angeles, CA, USA
| | - Lon S Schneider
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Debra Hawes
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Keith L Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA.
- Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars- Sinai Medical Center, Los Angeles, CA, USA.
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
6
|
Zeng HX, Qin SJ, Andersson J, Li SP, Zeng QG, Li JH, Wu QZ, Meng WJ, Oudin A, Kanninen KM, Jalava P, Dong GH, Zeng XW. The emerging roles of particulate matter-changed non-coding RNAs in the pathogenesis of Alzheimer's disease: A comprehensive in silico analysis and review. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 366:125440. [PMID: 39631655 DOI: 10.1016/j.envpol.2024.125440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 11/27/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
Research on epigenetic‒environmental interactions in the development of Alzheimer's disease (AD) has accelerated rapidly in recent decades. Numerous studies have demonstrated the contribution of ambient particulate matter (PM) to the onset of AD. Emerging evidence indicates that non-coding RNAs (ncRNAs), including long non-coding RNAs, circular RNAs, and microRNAs, play a role in the pathophysiology of AD. In this review, we provide an overview of PM-altered ncRNAs in the brain, with emphasis on their potential roles in the pathogenesis of AD. These results suggest that these PM-altered ncRNAs are involved in the regulation of amyloid-beta pathology, microtubule-associated protein Tau pathology, synaptic dysfunction, damage to the blood‒brain barrier, microglial dysfunction, dysmyelination, and neuronal loss. In addition, we utilized in silico analysis to explore the biological functions of PM-altered ncRNAs in the development of AD. This review summarizes the knowns and unknowns of PM-altered ncRNAs in AD pathogenesis and discusses the current dilemma regarding PM-altered ncRNAs as promising biomarkers of AD. Altogether, this is the first thorough review of the connection between PM exposure and ncRNAs in AD pathogenesis, which may offer novel insights into the prevention, diagnosis, and treatment of AD associated with ambient PM exposure.
Collapse
Affiliation(s)
- Hui-Xian Zeng
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shuang-Jian Qin
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | | | - Shen-Pan Li
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qing-Guo Zeng
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jia-Hui Li
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qi-Zhen Wu
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Wen-Jie Meng
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Anna Oudin
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Katja M Kanninen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Pasi Jalava
- Department of Environmental and Biological Science, University of Eastern Finland, Kuopio, Finland
| | - Guang-Hui Dong
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiao-Wen Zeng
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
7
|
Elhage A, Cohen S, Cummings J, van der Flier WM, Aisen P, Cho M, Bell J, Hampel H. Defining benefit: Clinically and biologically meaningful outcomes in the next-generation Alzheimer's disease clinical care pathway. Alzheimers Dement 2025; 21:e14425. [PMID: 39697158 PMCID: PMC11848336 DOI: 10.1002/alz.14425] [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: 07/22/2024] [Revised: 10/24/2024] [Accepted: 11/01/2024] [Indexed: 12/20/2024]
Abstract
To understand the potential benefits of emerging Alzheimer's disease (AD) therapies within and beyond clinical trial settings, there is a need to advance current outcome measurements into meaningful information relevant to all stakeholders. The relationship between the impact on disease biology and clinically measurable outcomes in cognition, function, and behavior must be considered when defining the meaningful benefit of early AD therapies. In this review, we discuss: (1) the lack of consideration for biomarkers in the current concept of meaningfulness in AD; (2) the lack of gold standards for determining minimal biologically and clinically important differences (MBCIDs) in AD trials; (3) how the treatment benefits of disease-modifying treatments are cumulative and increase over time; and (4) the different concepts of meaningfulness among key stakeholders. This review utilizes the future clinical biological framework of AD and aims to further integrate and expand the parameters of meaningful benefits toward a precision medicine framework. HIGHLIGHTS: Definition of meaningful benefit from Alzheimer's disease (AD) treatment varies across disease stage and stakeholder perspectives. Observable and meaningful outcomes must consider the clinical-biological nature of AD. Statistically significant effects or outcomes do not always equate to clinically meaningfulness. Assessment tools must reflect stage-specific subtle changes following treatment. Real-world evidence will support consensus, definition, and interpretation of clinical meaningfulness.
Collapse
Affiliation(s)
| | | | | | - Wiesje M. van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMCAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamThe Netherlands
- Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMCAmsterdamThe Netherlands
| | - Paul Aisen
- Alzheimer's Therapeutic Research Institute, University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Min Cho
- Eisai Inc.NutleyNew JerseyUSA
| | | | | |
Collapse
|
8
|
Rabinovici GD, Knopman DS, Arbizu J, Benzinger TLS, Donohoe KJ, Hansson O, Herscovitch P, Kuo PH, Lingler JH, Minoshima S, Murray ME, Price JC, Salloway SP, Weber CJ, Carrillo MC, Johnson KA. Updated Appropriate Use Criteria for Amyloid and Tau PET: A Report from the Alzheimer's Association and Society for Nuclear Medicine and Molecular Imaging Workgroup. J Nucl Med 2025:jnumed.124.268756. [PMID: 39778970 DOI: 10.2967/jnumed.124.268756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 01/11/2025] Open
Abstract
The Alzheimer's Association and the Society of Nuclear Medicine and Molecular Imaging convened a multidisciplinary workgroup to update appropriate use criteria (AUC) for amyloid positron emission tomography (PET) and to develop AUC for tau PET. Methods: The workgroup identified key research questions that guided a systematic literature review on clinical amyloid/tau PET. Building on this review, the workgroup developed 17 clinical scenarios in which amyloid or tau PET may be considered. A modified Delphi approach was used to rate each scenario by consensus as "rarely appropriate," "uncertain," or "appropriate." Ratings were performed separately for amyloid and tau PET as stand-alone modalities. Results: For amyloid PET, 7 scenarios were rated as appropriate, 2 as uncertain, and 8 as rarely appropriate. For tau PET, 5 scenarios were rated as appropriate, 6 as uncertain, and 6 as rarely appropriate. Conclusion: AUC for amyloid and tau PET provide expert recommendations for clinical use of these technologies in the evolving landscape of diagnostics and therapeutics for Alzheimer's disease.
Collapse
Affiliation(s)
- Gil D Rabinovici
- Department of Neurology and Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California;
| | - David S Knopman
- Mayo Clinic Neurology and Neurosurgery, Rochester, Minnesota
| | - Javier Arbizu
- Department of Nuclear Medicine, University of Navarra Clinic, Pamplona, Spain
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri; Knight Alzheimer's Disease Research Center, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Kevin J Donohoe
- Nuclear Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Peter Herscovitch
- Positron Emission Tomography Department, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Phillip H Kuo
- Medical Imaging, Medicine, and Biomedical Engineering, University of Arizona, Tucson, Arizona
| | - Jennifer H Lingler
- Department of Health and Community Systems, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Satoshi Minoshima
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah
| | | | - Julie C Price
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Stephen P Salloway
- Department of Neurology and Psychiatry the Warren Alpert School of Medicine, Brown University, Providence, Rhode Island
- Butler Hospital Memory and Aging Program, Providence, Rhode Island
| | | | | | - Keith A Johnson
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
- Molecular Neuroimaging, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts; and
- Departments of Neurology and Radiology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
9
|
Gong L, Liu D, Zhang B, Yu S, Xi C. Sex-Specific Entorhinal Cortex Functional Connectivity in Cognitively Normal Older Adults with Amyloid-β Pathology. Mol Neurobiol 2025; 62:475-484. [PMID: 38867110 PMCID: PMC11711718 DOI: 10.1007/s12035-024-04243-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 05/06/2024] [Indexed: 06/14/2024]
Abstract
Sex and apolipoprotein E (APOE) genotype have been shown to influence the risk and progression of Alzheimer's disease (AD). However, the impact of these factors on the functional connectivity of the entorhinal cortex (ERC) in clinically unpaired older adults (CUOA) with amyloid-β (Aβ +) pathology remains unclear. A total of 1022 cognitively normal older adults with Aβ + (603 females and 586 APOE ε4 +) from the Anti-Amyloid Treatment in Asymptomatic Alzheimer's (A4) study were included in this study. The 2 × 2 (gender, 2 APOE genotypes) analysis of covariance was performed to compare the demographic information, cognitive performance, and volumetric MRI data among these groups. Voxel-wise comparisons of bilateral ERC functional connectivity (FC) were conducted, and partial correlation analyses were used to explore the associations between cognitive performance and ERC-FC strength. We found that the APOE genotype influenced ERC functional connectivity mainly in the sensorimotor network (SMN). Males exhibited higher ERC-FC in the salience network (SN), while females displayed higher ERC-FC in the default mode network (DMN), executive control network (ECN), and reward network. The interplay of sex and APOE genotype on ERC-FC was observed in the SMN and cerebellar lobe. The ERC-FC was associated with executive function and memory performance in individuals with CUOA-Aβ + . Our findings provide evidence of sex-specific ERC functional connectivity compensation mechanism in cognitively normal older adults with Aβ + pathology. This study may contribute to a better understanding of the mechanisms underlying the early stages of AD and may help develop personalized interventions in preclinical AD.
Collapse
Affiliation(s)
- Liang Gong
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, 610017, Sichuan, China
| | - Duan Liu
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, 610017, Sichuan, China
| | - Bei Zhang
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, 610017, Sichuan, China
| | - Siyi Yu
- Department of Acupuncture & Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China.
| | - Chunhua Xi
- Department of Neurology, The Third Affiliated Hospital of Anhui Medical University, Huaihe Road 390, Heifei, 230061, Anhui, China.
| |
Collapse
|
10
|
Rabinovici GD, Knopman DS, Arbizu J, Benzinger TLS, Donohoe KJ, Hansson O, Herscovitch P, Kuo PH, Lingler JH, Minoshima S, Murray ME, Price JC, Salloway SP, Weber CJ, Carrillo MC, Johnson KA. Updated appropriate use criteria for amyloid and tau PET: A report from the Alzheimer's Association and Society for Nuclear Medicine and Molecular Imaging Workgroup. Alzheimers Dement 2025; 21:e14338. [PMID: 39776249 PMCID: PMC11772739 DOI: 10.1002/alz.14338] [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: 07/19/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION The Alzheimer's Association and the Society of Nuclear Medicine and Molecular Imaging convened a multidisciplinary workgroup to update appropriate use criteria (AUC) for amyloid positron emission tomography (PET) and to develop AUC for tau PET. METHODS The workgroup identified key research questions that guided a systematic literature review on clinical amyloid/tau PET. Building on this review, the workgroup developed 17 clinical scenarios in which amyloid or tau PET may be considered. A modified Delphi approach was used to rate each scenario by consensus as "rarely appropriate," "uncertain," or "appropriate." Ratings were performed separately for amyloid and tau PET as stand-alone modalities. RESULTS For amyloid PET, seven scenarios were rated as appropriate, two as uncertain, and eight as rarely appropriate. For tau PET, five scenarios were rated as appropriate, six as uncertain, and six as rarely appropriate. DISCUSSION AUC for amyloid and tau PET provide expert recommendations for clinical use of these technologies in the evolving landscape of diagnostics and therapeutics for Alzheimer's disease. HIGHLIGHTS A multidisciplinary workgroup convened by the Alzheimer's Association and the Society of Nuclear Medicine and Molecular Imaging updated the appropriate use criteria (AUC) for amyloid positron emission tomography (PET) and to develop AUC for tau PET. The goal of these updated AUC is to assist clinicians in identifying clinical scenarios in which amyloid or tau PET may be useful for guiding the diagnosis and management of patients who have, or are at risk for, cognitive decline These updated AUC are intended for dementia specialists who spend a significant proportion of their clinical effort caring for patients with cognitive complaints, as well as serve as a general reference for a broader audience interested in implementation of amyloid and tau PET in clinical practice.
Collapse
Affiliation(s)
- Gil D. Rabinovici
- Department of Neurology and Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | | | - Javier Arbizu
- Department of Nuclear MedicineUniversity of Navarra ClinicPamplonaSpain
| | - Tammie L. S. Benzinger
- Mallinckrodt Institute of RadiologyWashington University in St. Louis School of MedicineSt. LouisMissouriUSA
- Knight Alzheimer's Disease Research CenterWashington University in St. Louis School of MedicineSt. LouisMissouriUSA
| | - Kevin J. Donohoe
- Nuclear Medicine, Beth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Oskar Hansson
- Department of Clinical Sciences MalmöClinical Memory Research UnitFaculty of MedicineLund UniversityLundSweden
- Memory Clinic, Skåne University HospitalSkånes universitetssjukhusMalmöSweden
| | - Peter Herscovitch
- Positron Emission Tomography DepartmentNational Institutes of Health Clinical CenterBethesdaMarylandUSA
| | - Phillip H. Kuo
- Medical Imaging, Medicine, and Biomedical EngineeringUniversity of ArizonaTucsonArizonaUSA
| | - Jennifer H. Lingler
- Department of Health and Community SystemsUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Satoshi Minoshima
- Department of Radiology and Imaging SciencesUniversity of UtahSalt Lake CityUtahUSA
| | | | - Julie C. Price
- Department of RadiologyMassachusetts General Hospital, BostonCharlestownMassachusettsUSA
| | - Stephen P. Salloway
- Department of Neurology and Psychiatry the Warren Alpert School of Medicine at Brown UniversityProvidenceRhode IslandUSA
- Butler Hospital Memory and Aging ProgramProvidenceRhode IslandUSA
| | | | - Maria C. Carrillo
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
| | - Keith A. Johnson
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Molecular Neuroimaging, Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
- Departments of Neurology and RadiologyMassachusetts General HospitalBostonMassachusettsUSA
| |
Collapse
|
11
|
Testo AA, Roundy G, Dumas JA. Cognitive Decline in Alzheimer's Disease. Curr Top Behav Neurosci 2025; 69:181-195. [PMID: 39485649 DOI: 10.1007/7854_2024_527] [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: 11/03/2024]
Abstract
Deficits in memory, language, and other cognitive domains that impact an individual's ability to perform necessary tasks of daily living are symptoms of dementia, which is a major cause of death and disability in older adults. As the global population continues to age, deepening our understanding of dementia is crucial. Alzheimer's disease is the leading cause of dementia and accounts for between 60% and 80% of total dementia cases. Declines in episodic memory are considered a hallmark of Alzheimer's disease and occur early in disease progression. The cognitive effects of Alzheimer's disease differ from the cognitive changes expected in nonpathological or normal aging. While some cognitive changes are expected as a part of the aging processes, the declines in cognition associated with Alzheimer's disease are to a degree that the individual diagnosed with the disease is unable to function independently in activities of daily living. In this review, we will discuss how cognition is impacted by both normal and pathological aging, with a focus on Alzheimer's disease. We describe the progressive nature of Alzheimer's disease, as well as the effects of each stage of the disease on cognition.
Collapse
Affiliation(s)
- Abigail A Testo
- Department of Psychiatry, University of Vermont, Burlington, VT, USA
| | - Gwenyth Roundy
- Department of Psychiatry, University of Vermont, Burlington, VT, USA
| | - Julie A Dumas
- Department of Psychiatry, University of Vermont, Burlington, VT, USA.
| |
Collapse
|
12
|
Terao CM, Alexander MW, Chalmers RP, Boshmaf SZ, Paterson J, Black SE, Papp KV, Sperling RA, Rabin JS. Identifying cognitive test scores associated with early tau burden in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2025; 17:e70052. [PMID: 39811702 PMCID: PMC11730071 DOI: 10.1002/dad2.70052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 01/16/2025]
Abstract
INTRODUCTION This study aimed to identify cognitive tests that optimally relate to tau positron emission tomography (PET) signal in the inferior temporal cortex (ITC), a neocortical region associated with early tau accumulation in Alzheimer's disease (AD). METHODS We analyzed cross-sectional data from the harvard aging brain study (HABS) (n = 128) and the Anti-Amyloid Treatment in Asymptomatic Alzheimer's (A4) study (n = 393). We used elastic net regression to identify the most robust cognitive correlates of tau PET signal in the ITC. Secondary analyses examined whether the cognitive correlates remained significantly associated with tau after adjusting for structural brain measures. RESULTS Episodic memory measures, including both total and "process" scores, were the most robust correlates of ITC tau across both cohorts. These cognitive test scores remained significant after accounting for structural brain measures. DISCUSSION These findings highlight the potential of specific episodic memory test scores to detect and monitor neuropathological changes associated with early AD. Highlights Machine learning identified cognitive correlates of early Alzheimer's disease tau burden.Both traditional and process scores predicted early tau burden.Episodic memory scores were among the strongest correlates.Cognitive scores remained significant after accounting for structural brain measures.
Collapse
Affiliation(s)
- Caitlin M. Terao
- Hurvitz Brain Sciences ProgramSunnybrook Research InstituteTorontoOntarioCanada
- Department of Psychology, York UniversityTorontoOntarioCanada
| | - Madeline Wood Alexander
- Hurvitz Brain Sciences ProgramSunnybrook Research InstituteTorontoOntarioCanada
- Rehabilitation Sciences InstituteUniversity of TorontoTorontoOntarioCanada
| | | | - Silina Z. Boshmaf
- Hurvitz Brain Sciences ProgramSunnybrook Research InstituteTorontoOntarioCanada
| | - Jane Paterson
- Hurvitz Brain Sciences ProgramSunnybrook Research InstituteTorontoOntarioCanada
| | - Sandra E. Black
- Hurvitz Brain Sciences ProgramSunnybrook Research InstituteTorontoOntarioCanada
- Rehabilitation Sciences InstituteUniversity of TorontoTorontoOntarioCanada
- Division of NeurologyDepartment of MedicineSunnybrook Health Sciences CentreUniversity of TorontoTorontoOntarioCanada
| | - Kathryn V. Papp
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Reisa A. Sperling
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Jennifer S. Rabin
- Hurvitz Brain Sciences ProgramSunnybrook Research InstituteTorontoOntarioCanada
- Rehabilitation Sciences InstituteUniversity of TorontoTorontoOntarioCanada
- Division of NeurologyDepartment of MedicineSunnybrook Health Sciences CentreUniversity of TorontoTorontoOntarioCanada
- Harquail Centre for NeuromodulationSunnybrook Research InstituteTorontoOntarioCanada
| |
Collapse
|
13
|
Llorente-Saguer I, Oxtoby NP. A data-driven framework for biomarker discovery applied to optimizing modern clinical and preclinical trials on Alzheimer's disease. Brain Commun 2024; 6:fcae438. [PMID: 39664778 PMCID: PMC11632366 DOI: 10.1093/braincomms/fcae438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 11/07/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024] Open
Abstract
PET is used to measure tau protein accumulation in Alzheimer's disease. Multiple biomarkers have been proposed to track disease progression, most notably the standardized uptake value ratio of PET tracer uptake in a target region of interest relative to a reference region, but literature suggests these region choices are nontrivial. This study presents and evaluates a novel framework, BioDisCVR, designed to facilitate the discovery of useful biomarkers, demonstrated on [18F]AV-1451 tau PET data in multiple cohorts. BioDisCVR enhances signal-to-noise by conducting a data-driven search through the space of possible combinations of regional tau PET signals into a ratio of two composite regions, driven by a user-defined fitness function. This study compares ratio-based biomarkers discovered by the framework with state-of-the-art standardized uptake value ratio biomarkers. Data used is tau PET regional measurements from 198 individuals from the Alzheimer's Disease Neuroimaging Initiative database, used for discovery, and 42 from the Mayo Clinic Alzheimer's Disease Research Center and Mayo Clinic Study of Aging (MCSA), used for external validation. Biomarkers are evaluated by calculating clinical trial sample size estimates for 80% power and 20% effect size. Secondary metrics are a measure of longitudinal consistency (standard deviation of linear mixed-effects model residuals), and separation between cognitive groups (t-statistic of the change over time due to being cognitively impaired). When applied to preclinical (secondary prevention with CU individuals) and clinical (treatment aimed at cognitively impaired individuals) trials on Alzheimer's disease, our data-driven framework BioDisCVR discovered ratio-based tau PET biomarkers vastly superior to previous work, both reducing measurement error and sample size estimates for hypothetical clinical trials. Our analysis suggests remarkable potential for patient benefit (reduced exposure to health risks associated with experimental drugs) and substantial cost savings, through accelerated trials and reduced sample sizes. Our study supports the leveraging of data-driven methods like BioDisCVR for clinical benefit, with the potential to positively impact drug development in Alzheimer's disease and beyond.
Collapse
Affiliation(s)
- Isaac Llorente-Saguer
- UCL Hawkes Institute and Department of Medical Physics and Biomedical Engineering, University College London, London WC1E 6BT, UK
| | - Neil P Oxtoby
- UCL Hawkes Institute and Department of Computer Science, University College London, London WC1E 6BT, UK
| |
Collapse
|
14
|
Li JS, Tun SM, Ficek-Tani B, Xu W, Wang S, Horien CL, Toyonaga T, Nuli SS, Zeiss CJ, Powers AR, Zhao Y, Mormino EC, Fredericks CA. Medial Amygdalar Tau Is Associated With Mood Symptoms in Preclinical Alzheimer's Disease. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2024; 9:1301-1311. [PMID: 39059466 PMCID: PMC11625605 DOI: 10.1016/j.bpsc.2024.07.012] [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: 04/29/2024] [Revised: 07/01/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND While the amygdala receives early tau deposition in Alzheimer's disease (AD) and is involved in social and emotional processing, the relationship between amygdalar tau and early neuropsychiatric symptoms in AD is unknown. We sought to determine whether focal tau binding in the amygdala and abnormal amygdalar connectivity were detectable in a preclinical AD cohort and identify relationships between these and self-reported mood symptoms. METHODS We examined 598 individuals (347 amyloid positive [58% female], 251 amyloid negative [62% female] subset in tau positron emission tomography and functional magnetic resonance imaging cohorts) from the A4 (Anti-Amyloid Treatment in Asymptomatic AD) Study. In the tau positron emission tomography cohort, we used amygdalar segmentations to examine representative nuclei from 3 functional divisions of the amygdala. We analyzed between-group differences in division-specific tau binding in the amygdala in preclinical AD. We conducted seed-based functional connectivity analyses from each division in the functional magnetic resonance imaging cohort. Finally, we conducted exploratory post hoc correlation analyses between neuroimaging biomarkers of interest and anxiety and depression scores. RESULTS Amyloid-positive individuals demonstrated increased tau binding in the medial and lateral amygdala, and tau binding in these regions was associated with mood symptoms. Across amygdalar divisions, amyloid-positive individuals had relatively higher regional connectivity from the amygdala to other temporal regions, the insula, and the orbitofrontal cortex, but medial amygdala to retrosplenial cortex connectivity was lower. Medial amygdala to retrosplenial connectivity was negatively associated with anxiety symptoms, as was retrosplenial tau. CONCLUSIONS Our findings suggest that preclinical tau deposition in the amygdala and associated changes in functional connectivity may be related to early mood symptoms in AD.
Collapse
Affiliation(s)
- Joyce S Li
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Samantha M Tun
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | | | - Wanwan Xu
- Department of Biostatistics, Yale School of Medicine, New Haven, Connecticut
| | - Selena Wang
- Department of Biostatistics, Yale School of Medicine, New Haven, Connecticut
| | | | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | | | - Caroline J Zeiss
- Department of Comparative Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Albert R Powers
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
| | - Yize Zhao
- Department of Biostatistics, Yale School of Medicine, New Haven, Connecticut
| | - Elizabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | | |
Collapse
|
15
|
Mace RA, Law ME, Cohen JE, Ritchie CS, Okereke OI, Hoeppner BB, Brewer JA, Bartels SJ, Vranceanu AM. A Mindfulness-Based Lifestyle Intervention for Dementia Risk Reduction: Protocol for the My Healthy Brain Feasibility Randomized Controlled Trial. JMIR Res Protoc 2024; 13:e64149. [PMID: 39571150 PMCID: PMC11621724 DOI: 10.2196/64149] [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: 07/19/2024] [Revised: 08/29/2024] [Accepted: 09/28/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Lifestyle behavior change and mindfulness have direct and synergistic effects on cognitive functioning and may prevent Alzheimer disease and Alzheimer disease-related dementias (AD/ADRD). We are iteratively developing and testing My Healthy Brain (MHB), the first mindfulness-based lifestyle group program targeting AD/ADRD risk factors in older adults with subjective cognitive decline. Our pilot studies (National Institutes of Health [NIH] stage 1A) have shown that MHB is feasible, acceptable, and associated with improvement in lifestyle behavior and cognitive outcomes. OBJECTIVE We will compare the feasibility of MHB versus an education control (health enhancement program [HEP]) in 50 older adults (aged ≥60 y) with subjective cognitive decline and AD/ADRD risk factors. In an NIH stage 1B randomized controlled trial (RCT), we will evaluate feasibility benchmarks, improvements in cognitive and lifestyle outcomes, and engagement of hypothesized mechanisms. METHODS We are recruiting through clinics, flyers, web-based research platforms, and community partnerships. Participants are randomized to MHB or the HEP, both delivered in telehealth groups over 8 weeks. MHB participants learn behavior modification and mindfulness skills to achieve individualized lifestyle goals. HEP participants receive lifestyle education and group support. Assessments are repeated after the intervention and at a 6-month follow-up. Our primary outcomes are feasibility, acceptability, appropriateness, credibility, satisfaction, and fidelity benchmarks. The secondary outcomes are cognitive function and lifestyle (physical activity, sleep, nutrition, alcohol and tobacco use, and mental and social activity) behaviors. Data analyses will include the proportion of MHB and HEP participants who meet each benchmark (primary outcome) and paired samples 2-tailed t tests, Cohen d effect sizes, and the minimal clinically important difference for each measure (secondary outcomes). RESULTS Recruitment began in January 2024. We received 225 inquiries. Of these 225 individuals, 40 (17.8%) were eligible. Of the 40 eligible participants, 21 (52.5%) were enrolled in 2 group cohorts, 17 (42.5%) were on hold for future group cohorts, and 2 (5%) withdrew before enrollment. All participants have completed before the intervention assessments. All cohort 1 participants (9/21, 43%) have completed either MHB or the HEP (≥6 of 8 sessions) and after the intervention assessments. The intervention for cohort 2 (12/21, 57%) is ongoing. Adherence rates for the Garmin Vivosmart 5 (128/147, 87.1% weeks) and daily surveys (105/122, 86.1% weeks) are high. No enrolled participants have dropped out. Enrollment is projected to be completed by December 2024. CONCLUSIONS The RCT will inform the development of a larger efficacy RCT (NIH stage 2) of MHB versus the HEP in a more diverse sample of older adults, testing mechanisms of improvements through theoretically driven mediators and moderators. The integration of mindfulness with lifestyle behavior change in MHB has the potential to be an effective and sustainable approach for increasing the uptake of AD/ADRD risk reduction strategies among older adults. TRIAL REGISTRATION ClinicalTrials.gov NCT05934136; https://www.clinicaltrials.gov/study/NCT05934136. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/64149.
Collapse
Affiliation(s)
- Ryan A Mace
- Center for Health Outcomes and Interdisciplinary Research (CHOIR), Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Makenna E Law
- Center for Health Outcomes and Interdisciplinary Research (CHOIR), Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
| | - Joshua E Cohen
- Center for Health Outcomes and Interdisciplinary Research (CHOIR), Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
| | - Christine S Ritchie
- Harvard Medical School, Boston, MA, United States
- Mongan Institute Center for Aging and Serious Illness, Division of Palliative Care and Geriatric Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Olivia I Okereke
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
| | - Bettina B Hoeppner
- Harvard Medical School, Boston, MA, United States
- Health through Flourishing (HtF) Program, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
| | - Judson A Brewer
- Mindfulness Center, Brown University School of Public Health, Providence, MA, United States
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, MA, United States
| | - Stephen J Bartels
- Harvard Medical School, Boston, MA, United States
- Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
- The Mongan Institute, Massachusetts General Hospital, Boston, MA, United States
| | - Ana-Maria Vranceanu
- Center for Health Outcomes and Interdisciplinary Research (CHOIR), Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| |
Collapse
|
16
|
Budamagunta MS, Mori H, Silk J, Slez RR, Bognár B, Mendiola UR, Kálai T, Maezawa I, Voss JC. Nitroxyl Hybrids with Curcumin and Stilbene Scaffolds Display Potent Antioxidant Activity, Remodel the Amyloid Beta Oligomer, and Reverse Amyloid Beta-Induced Cytotoxicity. Antioxidants (Basel) 2024; 13:1411. [PMID: 39594552 PMCID: PMC11591036 DOI: 10.3390/antiox13111411] [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: 10/11/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
The disorder and heterogeneity of low-molecular-weight amyloid-beta oligomers (AβOs) underlie their participation in multiple modes of cellular dysfunction associated with the etiology of Alzheimer's disease (AD). The lack of specified conformational states in these species complicates efforts to select or design small molecules to targeting discrete pathogenic states. Furthermore, targeting AβOs alone may be therapeutically insufficient, as AD progresses as a multifactorial, self-amplifying cascade. To address these challenges, we have screened the activity of seven new candidates that serve as Paramagnetic Amyloid Ligand (PAL) candidates. PALs are bifunctional small molecules that both remodel the AβO structure and localize a potent antioxidant that mimics the activity of SOD within live cells. The candidates are built from either a stilbene or curcumin scaffold with nitroxyl moiety to serve as catalytic antioxidants. Measurements of PAL AβO binding and remolding along with assessments of bioactivity allow for the extraction of useful SAR information from screening data. One candidate (HO-4450; PMT-307), with a six-membered nitroxyl ring attached to a stilbene ring, displays the highest potency in protecting against cell-derived Aβ. A preliminary low-dose evaluation in AD model mice provides evidence of modest treatment effects by HO-4450. The results for the curcumin PALs demonstrate that the retention of the native curcumin phenolic groups is advantageous to the design of the hybrid PAL candidates. Finally, the PAL remodeling of AβO secondary structures shows a reasonable correlation between a candidate's bioactivity and its ability to reduce the fraction of antiparallel β-strand.
Collapse
Affiliation(s)
- Madhu S. Budamagunta
- Department of Biochemistry & Molecular Medicine, University of California, Davis, CA 95616, USA; (M.S.B.); (J.S.); (R.R.S.)
| | - Hidetoshi Mori
- Center for Genomic Pathology, University of California Davis, Sacramento, CA 95817, USA
| | - Joshua Silk
- Department of Biochemistry & Molecular Medicine, University of California, Davis, CA 95616, USA; (M.S.B.); (J.S.); (R.R.S.)
- Paramag Biosciences Inc., 720 Olive Drive, Davis, CA 95616, USA
| | - Ryan R. Slez
- Department of Biochemistry & Molecular Medicine, University of California, Davis, CA 95616, USA; (M.S.B.); (J.S.); (R.R.S.)
| | - Balázs Bognár
- Institute of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Pécs, Honvéd St. 1., H-7624 Pécs, Hungary; (B.B.); (T.K.)
- János Szentágothai Research Center, Ifjúság St. 20., H-7624 Pécs, Hungary
| | - Ulises Ruiz Mendiola
- M.I.N.D. Institute and Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA 95817, USA;
| | - Tamás Kálai
- Institute of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Pécs, Honvéd St. 1., H-7624 Pécs, Hungary; (B.B.); (T.K.)
- János Szentágothai Research Center, Ifjúság St. 20., H-7624 Pécs, Hungary
| | - Izumi Maezawa
- M.I.N.D. Institute and Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA 95817, USA;
| | - John C. Voss
- Department of Biochemistry & Molecular Medicine, University of California, Davis, CA 95616, USA; (M.S.B.); (J.S.); (R.R.S.)
- Paramag Biosciences Inc., 720 Olive Drive, Davis, CA 95616, USA
| |
Collapse
|
17
|
Jang H, Chun MY, Yun J, Kim JP, Kang SH, Weiner M, Kim HJ, Na DL, Hong C, Son SJ, Roh HW, Lee T, Lee E, Lee EH, Shin D, Ham H, Gu Y, Kim Y, Kim C, Woo S, Seo SW. Ethnic differences in the prevalence of amyloid positivity and cognitive trajectories. Alzheimers Dement 2024; 20:7556-7566. [PMID: 39315862 PMCID: PMC11567875 DOI: 10.1002/alz.14247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/03/2024] [Accepted: 08/02/2024] [Indexed: 09/25/2024]
Abstract
INTRODUCTION We investigated the prevalence of amyloid beta (Aβ) positivity (+) and cognitive trajectories in Koreans and non-Hispanic Whites (NHWs). METHODS We included 5121 Koreans from multiple centers across South Korea and 929 NHWs from the Alzheimer's Disease Neuroimaging Initiative (ADNI). Participants underwent Aβ positron emission tomography and were categorized into cognitively unimpaired (CU), mild cognitive impairment (MCI), and dementia stages. Age, sex, education, and apolipoprotein E. genotype were adjusted using multivariable logistic regression and stabilized inverse probability of treatment weights based on the propensity scores to mitigate imbalances in these variables. RESULTS The prevalence of Aβ+ was lower in CU Koreans than in CU NHWs (adjusted odds ratio 0.60). Aβ+ Koreans showed a faster cognitive decline than Aβ+ NHWs in the CU (B = -0.314, p = .004) and MCI stages (B = -0.385, p < .001). DISCUSSION Ethnic characteristics of Aβ biomarkers should be considered in research and clinical application of Aβ-targeted therapies in diverse populations. HIGHLIGHTS Koreans have a lower prevalence of Aβ positivity compared to NHWs in the CU stage. The effects of Alzheimer's risk factors on Aβ positivity differ between Koreans and NHWs. Aβ-positive (Aβ+) Koreans show faster cognitive decline than Aβ+ NHWs in the CU and MCI stages.
Collapse
Affiliation(s)
- Hyemin Jang
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
- Department of NeurologySeoul National University HospitalSeoul National University College of MedicineJongno‐guSeoulSouth Korea
| | - Min Young Chun
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
- Department of NeurologyYonsei University College of MedicineSeodaemun‐guSeoulSouth Korea
- Department of NeurologyYongin Severance HospitalYonsei University Health SystemYongin‐siGyeonggi‐doSouth Korea
| | - Jihwan Yun
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
- Department of NeurologySoonchunhyang University Bucheon HospitalBucheon‐siGyeonggi‐doSouth Korea
| | - Jun Pyo Kim
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
- Alzheimer's Disease Convergence Research CenterSamsung Medical CenterGangnam‐guSeoulSouth Korea
| | - Sung Hoon Kang
- Department of NeurologyKorea University Guro HospitalKorea University College of MedicineGuro‐guSeoulSouth Korea
| | - Michael Weiner
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Hee Jin Kim
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
- Alzheimer's Disease Convergence Research CenterSamsung Medical CenterGangnam‐guSeoulSouth Korea
- Department of Health Sciences and TechnologySAIHST, Sungkyunkwan UniversitySeoulSouth Korea
- Department of Digital HealthSAIHST, Sungkyunkwan UniversityGangnam‐guSeoulSouth Korea
| | - Duk L. Na
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
- Alzheimer's Disease Convergence Research CenterSamsung Medical CenterGangnam‐guSeoulSouth Korea
| | - Chang‐Hyung Hong
- Department of PsychiatryAjou University School of MedicineAjou University HospitalSuwon‐siGyeonggi‐doSouth Korea
| | - Sang Joon Son
- Department of PsychiatryAjou University School of MedicineAjou University HospitalSuwon‐siGyeonggi‐doSouth Korea
| | - Hyun Woong Roh
- Department of PsychiatryAjou University School of MedicineAjou University HospitalSuwon‐siGyeonggi‐doSouth Korea
| | - Tae‐Kyeong Lee
- Department of NeurologySoonchunhyang University Bucheon HospitalBucheon‐siGyeonggi‐doSouth Korea
| | - Eek‐Sung Lee
- Department of NeurologySoonchunhyang University Bucheon HospitalBucheon‐siGyeonggi‐doSouth Korea
| | - Eun Hye Lee
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
| | - Daeun Shin
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
| | - Hongki Ham
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
- Alzheimer's Disease Convergence Research CenterSamsung Medical CenterGangnam‐guSeoulSouth Korea
| | - Yuna Gu
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
- Department of Health Sciences and TechnologySAIHST, Sungkyunkwan UniversitySeoulSouth Korea
| | - Yeshin Kim
- Department of NeurologyKangwon National University School of MedicineGangwon‐doSouth Korea
| | - Chi‐Hun Kim
- Department of NeurologyHallym University Sacred Heart HospitalAnyang‐siGyeonggi‐doSouth Korea
| | - Sook‐young Woo
- Biomedical Statistics CenterData Science Research InstituteSamsung Medical CenterGangnam‐guSeoulSouth Korea
| | - Sang Won Seo
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
- Alzheimer's Disease Convergence Research CenterSamsung Medical CenterGangnam‐guSeoulSouth Korea
- Department of Health Sciences and TechnologySAIHST, Sungkyunkwan UniversitySeoulSouth Korea
- Department of Digital HealthSAIHST, Sungkyunkwan UniversityGangnam‐guSeoulSouth Korea
| | | |
Collapse
|
18
|
Beer S, Elmenhorst D, Bischof GN, Ramirez A, Bauer A, Drzezga A. Explainable artificial intelligence identifies an AQP4 polymorphism-based risk score associated with brain amyloid burden. Neurobiol Aging 2024; 143:19-29. [PMID: 39208715 DOI: 10.1016/j.neurobiolaging.2024.08.002] [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: 02/09/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
Aquaporin-4 (AQP4) is hypothesized to be a component of the glymphatic system, a pathway for removing brain interstitial solutes like amyloid-β (Aβ). Evidence exists that genetic variation of AQP4 impacts Aβ clearance, clinical outcome in Alzheimer's disease as well as sleep measures. We examined whether a risk score calculated from several AQP4 single-nucleotide polymorphisms (SNPs) is related to Aβ neuropathology in older cognitively unimpaired white individuals. We used a machine learning approach and explainable artificial intelligence to extract information on synergistic effects of AQP4 SNPs on brain amyloid burden from the ADNI cohort. From this information, we formulated a sex-specific AQP4 SNP-based risk score and evaluated it using data from the screening process of the A4 study. We found in both cohorts significant associations of the risk score with brain amyloid burden. The results support the hypothesis of an involvement of the glymphatic system, and particularly AQP4, in brain amyloid aggregation pathology. They suggest also that different AQP4 SNPs exert a synergistic effect on the build-up of brain amyloid burden.
Collapse
Affiliation(s)
- Simone Beer
- Institute of Neuroscience and Medicine (INM-2), Forschungszentrum Jülich, Germany.
| | - David Elmenhorst
- Institute of Neuroscience and Medicine (INM-2), Forschungszentrum Jülich, Germany; Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Gerard N Bischof
- Institute of Neuroscience and Medicine (INM-2), Forschungszentrum Jülich, Germany; Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Alfredo Ramirez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn-Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Department for Neurodegenerative Diseases and Geriatric Psychiatry, Bonn, Germany; Department of Psychiatry and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, TX, United States
| | - Andreas Bauer
- Institute of Neuroscience and Medicine (INM-2), Forschungszentrum Jülich, Germany
| | - Alexander Drzezga
- Institute of Neuroscience and Medicine (INM-2), Forschungszentrum Jülich, Germany; Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn-Cologne, Germany
| |
Collapse
|
19
|
Crawford JL, Berry AS. Examining resilience to Alzheimer's disease through the lens of monoaminergic neuromodulator systems. Trends Neurosci 2024; 47:892-903. [PMID: 39368845 PMCID: PMC11563896 DOI: 10.1016/j.tins.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/15/2024] [Accepted: 09/16/2024] [Indexed: 10/07/2024]
Abstract
The monoaminergic nuclei are thought to be some of the earliest sites of Alzheimer's disease (AD) pathology in the brain, with tau-containing pretangles appearing in these nuclei decades before the onset of clinical impairments. It has increasingly been recognized that monoamine systems represent a critical target of investigation towards understanding the progression of AD and designing early detection and treatment approaches. This review synthesizes evidence across animal studies, human neuropathology, and state-of-the-art neuroimaging and daily life assessment methods in humans, which demonstrate robust relationships between monoamine systems and AD pathophysiology and behavior. Further, the review highlights the promise of multimethod, multisystem approaches to studying monoaminergic mechanisms of resilience to AD pathology.
Collapse
Affiliation(s)
| | - Anne S Berry
- Department of Psychology, Brandeis University, Waltham, MA, USA.
| |
Collapse
|
20
|
Divya R, Shantha Selva Kumari R. Multi-instance learning attention model for amyloid quantification of brain sub regions in longitudinal cognitive decline. Brain Res 2024; 1842:149103. [PMID: 38955250 DOI: 10.1016/j.brainres.2024.149103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 05/21/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024]
Abstract
Amyloid PET scans help in identifying the beta-amyloid deposition in different brain regions. The purpose of this study is to develop a deep learning model that can automate the task of finding amyloid deposition in different regions of the brain only by using PET scan and without the corresponding MRI scan. 2647 18F-Florbetapir PET scans are collected from Alzheimer's Disease Neuroimaging Initiative (ADNI) from multiple centres taken over a period. A deep learning model based on multi-instance learning and attention is proposed which is trained and validated using 80% of the scans and the remaining 20% of the scans are used for testing the model. The performance of the model is validated using Mean Absolute Error (MAE) and Root Mean Squared Error (RMSE). The proposed model is further tested upon an external dataset consisting of 1413 18F-Florbetapir PET scans from the Anti-Amyloid Treatment in Asymptomatic Alzheimer's (A4) study. The proposed model achieves MAE of 0.0243 and RMSE of 0.0320 for summary Standardized Uptake Value Ratio (SUVR) based on composite reference region for ADNI test set. When tested on the A4-study dataset, the proposed model achieves MAE of 0.038 and RMSE of 0.0495 for summary SUVR based on the composite region. The results show that the proposed model provides less MAE and RMSE when compared with existing models. A graphical user interface is developed based on the proposed model where the predictions are made by selecting the files of 18F-Florbetapir PET scans.
Collapse
Affiliation(s)
- R Divya
- Department of Electronics and Communication Engineering, Mepco Schlenk Engineering College, Sivakasi 626 005, Tamil Nadu, India.
| | - R Shantha Selva Kumari
- Department of Electronics and Communication Engineering, Mepco Schlenk Engineering College, Sivakasi 626 005, Tamil Nadu, India.
| |
Collapse
|
21
|
Kazemeini S, Nadeem-Tariq A, Shih R, Rafanan J, Ghani N, Vida TA. From Plaques to Pathways in Alzheimer's Disease: The Mitochondrial-Neurovascular-Metabolic Hypothesis. Int J Mol Sci 2024; 25:11720. [PMID: 39519272 PMCID: PMC11546801 DOI: 10.3390/ijms252111720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) presents a public health challenge due to its progressive neurodegeneration, cognitive decline, and memory loss. The amyloid cascade hypothesis, which postulates that the accumulation of amyloid-beta (Aβ) peptides initiates a cascade leading to AD, has dominated research and therapeutic strategies. The failure of recent Aβ-targeted therapies to yield conclusive benefits necessitates further exploration of AD pathology. This review proposes the Mitochondrial-Neurovascular-Metabolic (MNM) hypothesis, which integrates mitochondrial dysfunction, impaired neurovascular regulation, and systemic metabolic disturbances as interrelated contributors to AD pathogenesis. Mitochondrial dysfunction, a hallmark of AD, leads to oxidative stress and bioenergetic failure. Concurrently, the breakdown of the blood-brain barrier (BBB) and impaired cerebral blood flow, which characterize neurovascular dysregulation, accelerate neurodegeneration. Metabolic disturbances such as glucose hypometabolism and insulin resistance further impair neuronal function and survival. This hypothesis highlights the interconnectedness of these pathways and suggests that therapeutic strategies targeting mitochondrial health, neurovascular integrity, and metabolic regulation may offer more effective interventions. The MNM hypothesis addresses these multifaceted aspects of AD, providing a comprehensive framework for understanding disease progression and developing novel therapeutic approaches. This approach paves the way for developing innovative therapeutic strategies that could significantly improve outcomes for millions affected worldwide.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas A. Vida
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA; (S.K.); (A.N.-T.); (R.S.); (J.R.); (N.G.)
| |
Collapse
|
22
|
Lee S, Zide BS, Palm ST, Drew WJ, Sperling RA, Jacobs HIL, Siddiqi SH, Donovan NJ. Specific Association of Worry With Amyloid-β But Not Tau in Cognitively Unimpaired Older Adults. Am J Geriatr Psychiatry 2024; 32:1203-1214. [PMID: 38763835 DOI: 10.1016/j.jagp.2024.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/21/2024]
Abstract
OBJECTIVE Anxiety disorders and subsyndromal anxiety symptoms are highly prevalent in late life. Recent studies support that anxiety may be a neuropsychiatric symptom during preclinical Alzheimer's disease (AD) and that higher anxiety is associated with more rapid cognitive decline and progression to cognitive impairment. However, the associations of specific anxiety symptoms with AD pathologies and with co-occurring subjective and objective cognitive changes have not yet been established. METHODS Baseline data from the A4 and Longitudinal Evaluation of Amyloid Risk and Neurodegeneration studies were analyzed. Older adult participants (n = 4,486) underwent assessments of anxiety (State-Trait Anxiety Inventory-6 item version [STAI]), and cerebral amyloid-beta (Aβ; 18F-florbetapir) PET and a subset underwent tau (18F-flortaucipir) PET. Linear regressions estimated associations of Aβ in a cortical composite and tau in the amygdala, entorhinal, and inferior temporal regions with STAI-Total and individual STAI item scores. Models adjusted for age, sex, education, marital status, depression, Apolipoprotein ε4 genotype, and subjective and objective cognition (Cognitive Function Index-participant; Preclinical Alzheimer Cognitive Composite). RESULTS Greater Aβ deposition was significantly associated with higher STAI-Worry, adjusting for all covariates, but not with other STAI items or STAI-Total scores. In mediation analyses, the association of Aβ with STAI-Worry was partially mediated by subjective cognition with a stronger direct effect. No associations were found for regional tau deposition with STAI-Total or STAI-Worry score. CONCLUSION Greater worry was associated with Aβ but not tau deposition, independent of subjective and objective cognition in cognitively unimpaired (CU) older adults. These findings implicate worry as an early, specific behavioral marker and a possible therapeutic target in preclinical AD.
Collapse
Affiliation(s)
- Soyoung Lee
- Division of Geriatric Psychiatry, Department of Psychiatry, Brigham and Women's Hospital (SL, BSZ, NJD), Harvard Medical School, Boston, MA; Center for Brain Circuit Therapeutics, Brigham and Women's Hospital (SL, STP, WJD, SHS), Boston, MA.
| | - Benjamin S Zide
- Division of Geriatric Psychiatry, Department of Psychiatry, Brigham and Women's Hospital (SL, BSZ, NJD), Harvard Medical School, Boston, MA
| | - Stephan T Palm
- Center for Brain Circuit Therapeutics, Brigham and Women's Hospital (SL, STP, WJD, SHS), Boston, MA; Department of Neurology, Brigham and Women's Hospital (STP, WJD, RAS, NJD), Harvard Medical School, Boston, MA
| | - William J Drew
- Center for Brain Circuit Therapeutics, Brigham and Women's Hospital (SL, STP, WJD, SHS), Boston, MA; Department of Neurology, Brigham and Women's Hospital (STP, WJD, RAS, NJD), Harvard Medical School, Boston, MA
| | - Reisa A Sperling
- Department of Neurology, Brigham and Women's Hospital (STP, WJD, RAS, NJD), Harvard Medical School, Boston, MA; Department of Neurology, Massachusetts General Hospital (RAS, NJD), Harvard Medical School, Boston, MA
| | - Heidi I L Jacobs
- Department of Radiology, Massachusetts General Hospital (HILJ), Harvard Medical School, Boston, MA; School for Mental Health and Neuroscience, Alzheimer Centre Limburg (HILJ), Maastricht University, Maastricht, The Netherlands
| | - Shan H Siddiqi
- Center for Brain Circuit Therapeutics, Brigham and Women's Hospital (SL, STP, WJD, SHS), Boston, MA; Department of Psychiatry, Brigham and Women's Hospital (SHS), Harvard Medical School, Boston, MA
| | - Nancy J Donovan
- Division of Geriatric Psychiatry, Department of Psychiatry, Brigham and Women's Hospital (SL, BSZ, NJD), Harvard Medical School, Boston, MA; Department of Neurology, Brigham and Women's Hospital (STP, WJD, RAS, NJD), Harvard Medical School, Boston, MA; Department of Psychiatry, Massachusetts General Hospital (NJD), Harvard Medical School, Boston, MA
| |
Collapse
|
23
|
Dogan S, Barua PD, Baygin M, Tuncer T, Tan RS, Ciaccio EJ, Fujita H, Devi A, Acharya UR. Lattice 123 pattern for automated Alzheimer's detection using EEG signal. Cogn Neurodyn 2024; 18:2503-2519. [PMID: 39555305 PMCID: PMC11564704 DOI: 10.1007/s11571-024-10104-1] [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: 10/16/2023] [Revised: 03/01/2024] [Accepted: 03/07/2024] [Indexed: 11/19/2024] Open
Abstract
This paper presents an innovative feature engineering framework based on lattice structures for the automated identification of Alzheimer's disease (AD) using electroencephalogram (EEG) signals. Inspired by the Shannon information entropy theorem, we apply a probabilistic function to create the novel Lattice123 pattern, generating two directed graphs with minimum and maximum distance-based kernels. Using these graphs and three kernel functions (signum, upper ternary, and lower ternary), we generate six feature vectors for each input signal block to extract textural features. Multilevel discrete wavelet transform (MDWT) was used to generate low-level wavelet subbands. Our proposed model mirrors deep learning approaches, facilitating feature extraction in frequency and spatial domains at various levels. We used iterative neighborhood component analysis to select the most discriminative features from the extracted vectors. An iterative hard majority voting and a greedy algorithm were used to generate voted vectors to select the optimal channel-wise and overall results. Our proposed model yielded a classification accuracy of more than 98% and a geometric mean of more than 96%. Our proposed Lattice123 pattern, dynamic graph generation, and MDWT-based multilevel feature extraction can detect AD accurately as the proposed pattern can extract subtle changes from the EEG signal accurately. Our prototype is ready to be validated using a large and diverse database.
Collapse
Affiliation(s)
- Sengul Dogan
- Department of Digital Forensics Engineering, College of Technology, Firat University, Elazig, Turkey
| | - Prabal Datta Barua
- School of Business (Information System), University of Southern Queensland, Springfield, Australia
| | - Mehmet Baygin
- Department of Computer Engineering, College of Engineering, Erzurum Technical University, Erzurum, Turkey
| | - Turker Tuncer
- Department of Digital Forensics Engineering, College of Technology, Firat University, Elazig, Turkey
| | - Ru-San Tan
- Department of Cardiology, National Heart Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Edward J. Ciaccio
- Department of Medicine, Columbia University Irving Medical Center, New York, NY USA
| | - Hamido Fujita
- Malaysia-Japan International Institute of Technology (MJIIT), Universiti Teknologi Malaysia, 54100 Kuala Lumpur, Malaysia
- Andalusian Research Institute in Data Science and Computational Intelligence, University of Granada, Granada, Spain
- Regional Research Center, Iwate Prefectural University, Iwate, Japan
| | - Aruna Devi
- School of Education and Tertiary Access, University of the Sunshine Coast, Sippy Downs, Caboolture Campus, QLD Australia
| | - U. Rajendra Acharya
- School of Mathematics, Physics and Computing, University of Southern Queensland, Springfield, Australia
- Centre for Health Research, University of Southern Queensland, Springfield, Australia
| |
Collapse
|
24
|
Davis MR, Robinson E, Koronyo Y, Salobrar-Garcia E, Rentsendorj A, Gaire BP, Mirzaei N, Kayed R, Sadun AA, Ljubimov AV, Schneider LS, Hawes D, Black KL, Fuchs DT, Koronyo-Hamaoui M. Retinal ganglion cell vulnerability to pathogenic tau in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.613293. [PMID: 39345568 PMCID: PMC11430098 DOI: 10.1101/2024.09.17.613293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Accumulation of pathological tau isoforms, especially hyperphosphorylated tau at serine 396 (pS396-tau) and tau oligomers, has been demonstrated in the retinas of patients with mild cognitive impairment (MCI) and Alzheimer's disease (AD). Previous studies have noted a decrease in retinal ganglion cells (RGCs) in AD patients, but the presence and impact of pathological tau isoforms in RGCs and RGC integrity, particularly in early AD stages, have not been explored. To investigate this, we examined retinal superior temporal cross-sections from 25 patients with MCI (due to AD) or AD dementia and 16 cognitively normal (CN) controls, matched for age and gender. We utilized the RGC marker ribonucleic acid binding protein with multiple splicing (RBPMS) and Nissl staining to assess neuronal density in the ganglion cell layer (GCL). Our study found that hypertrophic RGCs containing pS396-tau and T22-positive tau oligomers were more frequently observed in MCI and AD patients compared to CN subjects. Quantitative analyses indicated a decline in RGC integrity, with 46-55% and 55-56% reductions of RBPMS+ RGCs (P<0.01) and Nissl+ GCL neurons (P<0.01-0.001), respectively, in MCI and AD patients. This decrease in RGC count was accompanied by increases in necroptotic-like morphology and the cleaved caspase-3 apoptotic marker in RGCs of AD patients. Furthermore, there was a 2.1 to 3.1-fold increase (P<0.05-0.0001) in pS396-tau-laden RGCs in MCI and AD patients, with a greater abundance observed in individuals with higher Braak stages (V-VI), more severe clinical dementia ratings (CDR=3), and lower mini-mental state examination (MMSE) scores. Strong correlations were noted between the decline in RGCs and the total amount of retinal pS396-tau and pS396-tau+ RGCs, with pS396-tau+ RGC counts correlating significantly with brain neurofibrillary tangle scores (r= 0.71, P= 0.0001), Braak stage (r= 0.65, P= 0.0009), and MMSE scores (r= -0.76, P= 0.0004). These findings suggest that retinal tauopathy, characterized by pS396-tau and oligomeric tau in hypertrophic RGCs, is associated with and may contribute to RGC degeneration in AD. Future research should validate these findings in larger cohorts and explore noninvasive retinal imaging techniques that target tau pathology in RGCs to improve AD detection and monitor disease progression.
Collapse
Affiliation(s)
- Miyah R. Davis
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Edward Robinson
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Elena Salobrar-Garcia
- Institute of Ophthalmologic Research Ramón Castroviejo, Complutense University of Madrid, 28040 Madrid, Spain. Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain. Health Research Institute, Clinico San Carlos Hospital (IdISSC), 28040 Madrid, Spain
| | - Altan Rentsendorj
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Bhakta P. Gaire
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nazanin Mirzaei
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch at Galveston, TX, USA
- Departments of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Alfredo A. Sadun
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
- Doheny Eye Institute, Los Angeles, CA, USA
| | - Alexander V. Ljubimov
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Eye Program, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lon S. Schneider
- Alzheimer’s Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Debra Hawes
- Alzheimer’s Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Keith L. Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
25
|
Magana‐Ramirez CM, Irizarry‐Martinez G, Gillen DL, Grill JD. Reasons for undergoing amyloid imaging among diverse enrollees in the A4 study. Alzheimers Dement 2024; 20:6060-6069. [PMID: 39041310 PMCID: PMC11497770 DOI: 10.1002/alz.14077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Understanding attitudes toward participation among diverse preclinical Alzheimer's disease (AD) trial participants could yield insights to instruct future recruitment. METHODS Using data from the Anti-Amyloid Treatment in Asymptomatic AD (A4) Study, we examined differences among mutually exclusive racial and ethnic groups in views and perceptions of amyloid imaging (VPAI), a measure of motivations to undergo amyloid biomarker testing in the setting of preclinical AD. We used linear regression to quantify differences at baseline. RESULTS Compared to non-Hispanic or Latino (NH) White participants, Hispanic or Latino (3.52 points, 95% confidence interval [CI]: [2.61, 4.42]); NH Asian (2.97 points, 95% CI: [1.71, 4.22]); and NH Black participants (2.79 points, 95% CI: [1.96, 3.63]) participants demonstrated higher levels of endorsement of the VPAI items at baseline. DISCUSSION Differences may exist among participants from differing ethnic and racial groups in motivations to undergo biomarker testing in the setting of a preclinical AD trial. HIGHLIGHTS Representative samples in AD clinical trials are vital to result in generalizability. We assessed motivations to undergo amyloid imaging in a preclinical AD trial. Racial and ethnic minority groups showed higher endorsement of VPAI items. Differences were driven by perceived risk, plan/prepare, and curiosity domains. Few observations among racial and ethnic groups changed after biomarker disclosure.
Collapse
Affiliation(s)
- Christina M. Magana‐Ramirez
- Department of StatisticsDonald Bren School of Information and Computer Sciences, University of CaliforniaIrvineCaliforniaUSA
| | | | - Daniel L. Gillen
- Department of StatisticsDonald Bren School of Information and Computer Sciences, University of CaliforniaIrvineCaliforniaUSA
- Alzheimer’s Disease Research Center, University of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
| | - Joshua D. Grill
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Alzheimer’s Disease Research Center, University of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
- Department of Psychiatry and Human BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| |
Collapse
|
26
|
Wang X, Luo L, Zhao J, Guo X, Tao L, Zhang F, Liu X, Gao B, Luo Y. Associations between sleep duration trajectories and cognitive decline: A longitudinal cohort study in China. Arch Gerontol Geriatr 2024; 124:105445. [PMID: 38733919 DOI: 10.1016/j.archger.2024.105445] [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: 01/27/2024] [Revised: 03/24/2024] [Accepted: 04/13/2024] [Indexed: 05/13/2024]
Abstract
OBJECT The relationship between sleep duration trajectories and cognitive decline remains uncertain. This study aims to examine the connections between various patterns of sleep duration and cognitive function. METHODS Group-based trajectory modeling (GBTM) was employed to identify longitudinal trajectories of sleep duration over four-year follow-up period, while considering age, sex and nap duration as adjustments. Logistic regression was utilized to analyze the association between sleep trajectories and cognition, with odds ratios (OR) and 95 % confidence intervals (CI) reported. Subgroup analyses based on various demographic characteristics were conducted to explore potential differences in sleep trajectories and cognitive decline across different population subgroups. RESULTS A total of 5061 participants were followed for four years, and three sleep duration trajectories were identified: high increasing (n = 2101, 41.6 %), stable increasing (n = 2087, 40.7 %), and low decreasing (n = 873, 17.7 %). After adjustment for basic demographic information, health status, and baseline cognition, the high increasing trajectory was found to be associated with cognitive decline in terms of global cognition (OR:1.52,95 %CI:1.18-1.96), mental intactness (OR:1.36,95 %CI:1.07-1.73) and episodic memory (OR:1.33, 95 %CI:1.05-1.67), as compared to stable increasing trajectory. These associations were particularly prominent among the non-elderly population (≤65 years) and those without depressive symptoms. CONCLUSION This study suggests that both high increasing and low decreasing sleep duration trajectories are linked to cognitive decline, as compared to the stable increasing trajectory. Long-term attention to changes in sleep duration facilitates early prevention of cognitive decline.
Collapse
Affiliation(s)
- Xiaonan Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, PR China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, PR China
| | - Lili Luo
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, PR China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, PR China
| | - Jianxi Zhao
- School of Applied Science, Beijing Information Science and Technology University, Beijing, 100192, China
| | - Xiuhua Guo
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, PR China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, PR China
| | - Lixin Tao
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, PR China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, PR China
| | - Feng Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, PR China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, PR China
| | - Xiangtong Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, PR China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, PR China
| | - Bo Gao
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, PR China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, PR China
| | - Yanxia Luo
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, PR China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, PR China.
| |
Collapse
|
27
|
Jackson RJ, Hyman BT, Serrano-Pozo A. Multifaceted roles of APOE in Alzheimer disease. Nat Rev Neurol 2024; 20:457-474. [PMID: 38906999 DOI: 10.1038/s41582-024-00988-2] [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: 05/24/2024] [Indexed: 06/23/2024]
Abstract
For the past three decades, apolipoprotein E (APOE) has been known as the single greatest genetic modulator of sporadic Alzheimer disease (AD) risk, influencing both the average age of onset and the lifetime risk of developing AD. The APOEε4 allele significantly increases AD risk, whereas the ε2 allele is protective relative to the most common ε3 allele. However, large differences in effect size exist across ethnoracial groups that are likely to depend on both global genetic ancestry and local genetic ancestry, as well as gene-environment interactions. Although early studies linked APOE to amyloid-β - one of the two culprit aggregation-prone proteins that define AD - in the past decade, mounting work has associated APOE with other neurodegenerative proteinopathies and broader ageing-related brain changes, such as neuroinflammation, energy metabolism failure, loss of myelin integrity and increased blood-brain barrier permeability, with potential implications for longevity and resilience to pathological protein aggregates. Novel mouse models and other technological advances have also enabled a number of therapeutic approaches aimed at either attenuating the APOEε4-linked increased AD risk or enhancing the APOEε2-linked AD protection. This Review summarizes this progress and highlights areas for future research towards the development of APOE-directed therapeutics.
Collapse
Affiliation(s)
- Rosemary J Jackson
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.
| | - Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.
| |
Collapse
|
28
|
Seto M, Hohman TJ, Mormino EC, Papp KV, Amariglio RE, Rentz DM, Johnson KA, Schultz AP, Sperling RA, Buckley RF, Yang HS. Parental History of Memory Impairment and β-Amyloid in Cognitively Unimpaired Older Adults. JAMA Neurol 2024; 81:798-804. [PMID: 38884955 PMCID: PMC11184498 DOI: 10.1001/jamaneurol.2024.1763] [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: 01/16/2024] [Accepted: 04/19/2024] [Indexed: 06/18/2024]
Abstract
Importance Studies have suggested that maternal history of late-onset Alzheimer disease, but not paternal, predisposes individuals to higher brain β-amyloid (Aβ) burden, reduced brain metabolism, and lower gray matter volumes. Objective To characterize maternal vs paternal history of memory impairment in terms of brain Aβ-positron emission tomography (Aβ-PET) and baseline cognition among a large sample of cognitively unimpaired older adults. Design, Setting, and Participants This cross-sectional study leveraged data from 4413 individuals who were screened for the Anti-Amyloid Treatment in Asymptomatic Alzheimer (A4) study, a randomized clinical trial conducted across 67 sites in the US, Australia, Canada, and Japan aimed at Alzheimer disease prevention. Data were collected between April 2014 and December 2017 and analyzed from December 2022 to June 2023. Participants were cognitively unimpaired adults (Clinical Dementia Rating = 0 and/or Mini-Mental State Examination score ≥25) between the ages of 65 and 85 years who underwent PET imaging to assess cortical Aβ levels for trial eligibility. A total of 4492 participants were screened, and 79 missing data were excluded. Main Outcomes and Measures Demographic characteristics (eg, age, sex, education), apolipoprotein E genotyping, participant-reported parental history of memory impairment and parental age at symptom onset were collected as variables. Parental history was assessed in terms of continuous neocortical 18F-florbetapir Aβ-PET and the Preclinical Alzheimer Cognitive Composite. Results Of 4413 individuals (mean [SD] age, 71.27 [4.66] years, 2617 women [59.3%]), mean Aβ-PET was elevated in individuals with history of memory impairment in both parents (n = 455; mean [SD] standardized uptake value ratio [SUVR] = 1.12 [0.19]; Wilcoxon P = 1.1 × 10-5) and in those with only maternal history (n = 1772; mean [SD] SUVR = 1.10 [0.19]; Wilcoxon P = 2.70 × 10-5) compared with those with only paternal history (n = 632; mean [SD] SUVR = 1.08 [0.18]; Wilcoxon P = 1.1 × 10-5) or no family history (n = 1554; mean [SD] SUVR = 1.08 [0.19]; Wilcoxon P = 1.1 × 10-5). Paternal history of early-onset memory impairment (age <65 years) but not late-onset (age ≥65 years) was associated with elevated participant Aβ-PET (mean [SD] SUVR = 1.19 [0.21]; P = 3.00 × 10-6) in comparison with no paternal history (mean [SD] SUVR = 1.09 [0.19]) whereas maternal history was associated with elevated Aβ in both early-onset and late-onset groups. There was no association with cognition. Conclusions and Relevance In this study, maternal history (at any age) and paternal history of early-onset memory impairment were associated with Aβ burden among asymptomatic older individuals. Sex-specific parental history may help inform clinicians on likelihood of Aβ burden in offspring and help identify high-risk individuals at the earliest stages of disease for prevention.
Collapse
Affiliation(s)
- Mabel Seto
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Elizabeth C. Mormino
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California
| | - Kathryn V. Papp
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Rebecca E. Amariglio
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Dorene M. Rentz
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Keith A. Johnson
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Department of Radiology, Massachusetts General Hospital, Boston
| | - Aaron P. Schultz
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown
| | - Reisa A. Sperling
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Rachel F. Buckley
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Hyun-Sik Yang
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
29
|
Shi H, Mirzaei N, Koronyo Y, Davis MR, Robinson E, Braun GM, Jallow O, Rentsendorj A, Ramanujan VK, Fert-Bober J, Kramerov AA, Ljubimov AV, Schneider LS, Tourtellotte WG, Hawes D, Schneider JA, Black KL, Kayed R, Selenica MLB, Lee DC, Fuchs DT, Koronyo-Hamaoui M. Identification of retinal oligomeric, citrullinated, and other tau isoforms in early and advanced AD and relations to disease status. Acta Neuropathol 2024; 148:3. [PMID: 38980423 PMCID: PMC11233395 DOI: 10.1007/s00401-024-02760-8] [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: 02/29/2024] [Revised: 06/07/2024] [Accepted: 06/26/2024] [Indexed: 07/10/2024]
Abstract
This study investigates various pathological tau isoforms in the retina of individuals with early and advanced Alzheimer's disease (AD), exploring their connection with disease status. Retinal cross-sections from predefined superior-temporal and inferior-temporal subregions and corresponding brains from neuropathologically confirmed AD patients with a clinical diagnosis of either mild cognitive impairment (MCI) or dementia (n = 45) were compared with retinas from age- and sex-matched individuals with normal cognition (n = 30) and non-AD dementia (n = 4). Retinal tau isoforms, including tau tangles, paired helical filament of tau (PHF-tau), oligomeric-tau (Oligo-tau), hyperphosphorylated-tau (p-tau), and citrullinated-tau (Cit-tau), were stereologically analyzed by immunohistochemistry and Nanostring GeoMx digital spatial profiling, and correlated with clinical and neuropathological outcomes. Our data indicated significant increases in various AD-related pretangle tau isoforms, especially p-tau (AT8, 2.9-fold, pS396-tau, 2.6-fold), Cit-tau at arginine residue 209 (CitR209-tau; 4.1-fold), and Oligo-tau (T22+, 9.2-fold), as well as pretangle and mature tau tangle forms like MC-1-positive (1.8-fold) and PHF-tau (2.3-fold), in AD compared to control retinas. MCI retinas also exhibited substantial increases in Oligo-tau (5.2-fold), CitR209-tau (3.5-fold), and pS396-tau (2.2-fold). Nanostring GeoMx analysis confirmed elevated retinal p-tau at epitopes: Ser214 (2.3-fold), Ser396 (2.6-fold), Ser404 (2.4-fold), and Thr231 (1.8-fold), particularly in MCI patients. Strong associations were found between retinal tau isoforms versus brain pathology and cognitive status: a) retinal Oligo-tau vs. Braak stage, neurofibrillary tangles (NFTs), and CDR cognitive scores (ρ = 0.63-0.71), b) retinal PHF-tau vs. neuropil threads (NTs) and ABC scores (ρ = 0.69-0.71), and c) retinal pS396-tau vs. NTs, NFTs, and ABC scores (ρ = 0.67-0.74). Notably, retinal Oligo-tau strongly correlated with retinal Aβ42 and arterial Aβ40 forms (r = 0.76-0.86). Overall, this study identifies and quantifies diverse retinal tau isoforms in MCI and AD patients, underscoring their link to brain pathology and cognition. These findings advocate for further exploration of retinal tauopathy biomarkers to facilitate AD detection and monitoring via noninvasive retinal imaging.
Collapse
Affiliation(s)
- Haoshen Shi
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
| | - Nazanin Mirzaei
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
| | - Miyah R Davis
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
| | - Edward Robinson
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
| | - Gila M Braun
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
| | - Ousman Jallow
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
| | - Altan Rentsendorj
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
| | - V Krishnan Ramanujan
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Justyna Fert-Bober
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Andrei A Kramerov
- Eye Program, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Division of Applied Cell Biology and Physiology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alexander V Ljubimov
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
- Eye Program, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Division of Applied Cell Biology and Physiology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lon S Schneider
- Departments of Psychiatry and the Behavioral Sciences and Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Warren G Tourtellotte
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Division of Applied Cell Biology and Physiology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Debra Hawes
- Department of Pathology Program in Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Julie A Schneider
- Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Keith L Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Maj-Linda B Selenica
- Sanders-Brown Center On Aging, Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Daniel C Lee
- Sanders-Brown Center On Aging, Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., A6212, Los Angeles, CA, 90048, USA.
- Division of Applied Cell Biology and Physiology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
30
|
Bollinger RM, Chen SW, Krauss MJ, Keleman AA, Kehrer-Dunlap A, Kaesler M, Ances BM, Stark SL. The Association Between Postural Sway and Preclinical Alzheimer Disease Among Community-Dwelling Older Adults. J Gerontol A Biol Sci Med Sci 2024; 79:glae091. [PMID: 38554257 PMCID: PMC11167487 DOI: 10.1093/gerona/glae091] [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: 10/26/2023] [Indexed: 04/01/2024] Open
Abstract
BACKGROUND It is unknown whether older adults with preclinical Alzheimer disease (AD) experience changes in postural sway compared with those without preclinical AD. The purpose of this study was to understand the effect of dual tasking on standing balance, or postural sway, for people with and without preclinical AD. METHODS A cross-sectional analysis of baseline data from a longitudinal cohort study. Participants were cognitively normal older adults with and without preclinical AD. Postural sway (path length) was tested using a force plate under standard and dual task balance conditions. Dual task cost (DTC) was calculated to examine performance change in balance conditions. Logistic regression models were used to predict preclinical AD status as a function of DTC. RESULTS 203 participants (65 preclinical AD+) were included. DTC for path length was significantly greater for participants with preclinical AD (DTC path length mean difference 19.8, 95% CI 2.6-37.0, t(201) = 2.29, p = .024). Greater DTC was significantly associated with increased odds of having preclinical AD (adjusted odds ratio for a 20-unit increase in DTC 1.16, 95% CI 1.02-1.32). CONCLUSIONS Older adults with preclinical AD are more likely to demonstrate significantly greater DTC in postural sway than those without preclinical AD. Dual tasking should be integrated into balance and fall risk assessments and may inform early detection of preclinical AD.
Collapse
Affiliation(s)
- Rebecca M Bollinger
- Program in Occupational Therapy, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Szu-Wei Chen
- Program in Occupational Therapy, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Melissa J Krauss
- Program in Occupational Therapy, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Audrey A Keleman
- Program in Occupational Therapy, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Abigail Kehrer-Dunlap
- Program in Occupational Therapy, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Megan Kaesler
- Program in Occupational Therapy, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Beau M Ances
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Susan L Stark
- Program in Occupational Therapy, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
31
|
Jadick MF, Robinson T, Farrell ME, Klinger H, Buckley RF, Marshall GA, Vannini P, Rentz DM, Johnson KA, Sperling RA, Amariglio RE. Associations Between Self and Study Partner Report of Cognitive Decline With Regional Tau in a Multicohort Study. Neurology 2024; 102:e209447. [PMID: 38810211 PMCID: PMC11226320 DOI: 10.1212/wnl.0000000000209447] [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: 12/13/2023] [Accepted: 03/04/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Self-reported cognitive decline is an early behavioral manifestation of Alzheimer disease (AD) at the preclinical stage, often believed to precede concerns reported by a study partner. Previous work shows cross-sectional associations with β-amyloid (Aβ) status and self-reported and study partner-reported cognitive decline, but less is known about their associations with tau deposition, particularly among those with preclinical AD. METHODS This cross-sectional study included participants from the Anti-Amyloid Treatment in Asymptomatic AD/Longitudinal Evaluation of Amyloid Risk and Neurodegeneration studies (N = 444) and the Harvard Aging Brain Study and affiliated studies (N = 231), which resulted in a cognitively unimpaired (CU) sample of individuals with both nonelevated (Aβ-) and elevated Aβ (Aβ+). All participants and study partners completed the Cognitive Function Index (CFI). Two regional tau composites were derived by averaging flortaucipir PET uptake in the medial temporal lobe (MTL) and neocortex (NEO). Global Aβ PET was measured in Centiloids (CLs) with Aβ+ >26 CL. We conducted multiple linear regression analyses to test associations between tau PET and CFI, covarying for amyloid, age, sex, education, and cohort. We also controlled for objective cognitive performance, measured using the Preclinical Alzheimer Cognitive Composite (PACC). RESULTS Across 675 CU participants (age = 72.3 ± 6.6 years, female = 59%, Aβ+ = 60%), greater tau was associated with greater self-CFI (MTL: β = 0.28 [0.12, 0.44], p < 0.001, and NEO: β = 0.26 [0.09, 0.42], p = 0.002) and study partner CFI (MTL: β = 0.28 [0.14, 0.41], p < 0.001, and NEO: β = 0.31 [0.17, 0.44], p < 0.001). Significant associations between both CFI measures and MTL/NEO tau PET were driven by Aβ+. Continuous Aβ showed an independent effect on CFI in addition to MTL and NEO tau for both self-CFI and study partner CFI. Self-CFI (β = 0.01 [0.001, 0.02], p = 0.03), study partner CFI (β = 0.01 [0.003, 0.02], p = 0.01), and the PACC (β = -0.02 [-0.03, -0.01], p < 0.001) were independently associated with MTL tau, but for NEO tau, PACC (β = -0.02 [-0.03, -0.01], p < 0.001) and study partner report (β = 0.01 [0.004, 0.02], p = 0.002) were associated, but not self-CFI (β = 0.01 [-0.001, 0.02], p = 0.10). DISCUSSION Both self-report and study partner report showed associations with tau in addition to Aβ. Additionally, self-report and study partner report were associated with tau above and beyond performance on a neuropsychological composite. Stratification analyses by Aβ status indicate that associations between self-reported and study partner-reported cognitive concerns with regional tau are driven by those at the preclinical stage of AD, suggesting that both are useful to collect on the early AD continuum.
Collapse
Affiliation(s)
- Michalina F Jadick
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Talia Robinson
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Michelle E Farrell
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hannah Klinger
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Rachel F Buckley
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Gad A Marshall
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Patrizia Vannini
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Dorene M Rentz
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Keith A Johnson
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Reisa A Sperling
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Rebecca E Amariglio
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
32
|
Li JS, Tun SM, Ficek-Tani B, Xu W, Wang S, Horien CL, Toyonaga T, Nuli SS, Zeiss CJ, Powers AR, Zhao Y, Mormino EC, Fredericks CA. Medial amygdalar tau is associated with anxiety symptoms in preclinical Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.03.597160. [PMID: 38895308 PMCID: PMC11185761 DOI: 10.1101/2024.06.03.597160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
BACKGROUND While the amygdala receives early tau deposition in Alzheimer's disease (AD) and is involved in social and emotional processing, the relationship between amygdalar tau and early neuropsychiatric symptoms in AD is unknown. We sought to determine whether focal tau binding in the amygdala and abnormal amygdalar connectivity were detectable in a preclinical AD cohort and identify relationships between these and self-reported mood symptoms. METHODS We examined n=598 individuals (n=347 amyloid-positive (58% female), n=251 amyloid-negative (62% female); subset into tau PET and fMRI cohorts) from the A4 Study. In our tau PET cohort, we used amygdalar segmentations to examine representative nuclei from three functional divisions of the amygdala. We analyzed between-group differences in division-specific tau binding in the amygdala in preclinical AD. We conducted seed-based functional connectivity analyses from each division in the fMRI cohort. Finally, we conducted exploratory post-hoc correlation analyses between neuroimaging biomarkers of interest and anxiety and depression scores. RESULTS Amyloid-positive individuals demonstrated increased tau binding in medial and lateral amygdala (F(4,442)=14.61, p=0.00045; F(4,442)=5.83, p=0.024, respectively). Across amygdalar divisions, amyloid-positive individuals had relatively increased regional connectivity from amygdala to other temporal regions, insula, and orbitofrontal cortex. There was an interaction by amyloid group between tau binding in the medial and lateral amygdala and anxiety. Medial amygdala to retrosplenial connectivity negatively correlated with anxiety symptoms (rs=-0.103, p=0.015). CONCLUSIONS Our findings suggest that preclinical tau deposition in the amygdala may result in meaningful changes in functional connectivity which may predispose patients to mood symptoms.
Collapse
Affiliation(s)
- Joyce S Li
- Department of Neurology, Yale School of Medicine, New Haven, CT
| | - Samantha M Tun
- Department of Neurology, Yale School of Medicine, New Haven, CT
| | | | - Wanwan Xu
- Department of Biostatistics, Yale School of Medicine, New Haven, CT
| | - Selena Wang
- Department of Biostatistics, Yale School of Medicine, New Haven, CT
| | | | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT
| | | | - Caroline J Zeiss
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT
| | - Albert R Powers
- Department of Psychiatry, Yale School of Medicine, New Haven, CT
| | - Yize Zhao
- Department of Biostatistics, Yale School of Medicine, New Haven, CT
| | - Elizabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA
| | | |
Collapse
|
33
|
Lozupone M, Dibello V, Sardone R, Castellana F, Zupo R, Lampignano L, Bortone I, Stallone R, Altamura M, Bellomo A, Daniele A, Solfrizzi V, Panza F. Lessons learned from the failure of solanezumab as a prospective treatment strategy for Alzheimer's disease. Expert Opin Drug Discov 2024; 19:639-647. [PMID: 38685682 DOI: 10.1080/17460441.2024.2348142] [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: 02/19/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION In the last decade, the efforts conducted for discovering Alzheimer's Disease (AD) treatments targeting the best-known pathogenic factors [amyloid-β (Aβ), tau protein, and neuroinflammation] were mostly unsuccessful. Given that a systemic failure of Aβ clearance was supposed to primarily contribute to AD development and progression, disease-modifying therapies with anti-Aβ monoclonal antibodies (e.g. solanezumab, bapineuzumab, gantenerumab, aducanumab, lecanemab and donanemab) are ongoing in randomized clinical trials (RCTs) with contrasting results. AREAS COVERED The present Drug Discovery Case History analyzes the failures of RCTs of solanezumab on AD. Furthermore, the authors review the pharmacokinetics, pharmacodynamics, and tolerability effect of solanezumab from preclinical studies with its analogous m266 in mice. Finally, they describe the RCTs with cognitive, cerebrospinal fluid and neuroimaging findings in mild-to-moderate AD (EXPEDITION studies) and in secondary prevention studies (A4 and DIAN-TU). EXPERT OPINION Solanezumab was one of the first anti-Aβ monoclonal antibodies to be tested in preclinical and clinical AD showing to reduce brain Aβ level by acting on soluble monomeric form of Aβ peptide without significant results on deposits. Unfortunately, this compound showed to accelerate cognitive decline in both asymptomatic and symptomatic trial participants, and this failure of solanezumab further questioned the Aβ cascade hypothesis of AD.
Collapse
Affiliation(s)
- Madia Lozupone
- Department of Translational Biomedicine and Neuroscience "DiBraiN", University of Bari Aldo Moro, Bari, Italy
| | - Vittorio Dibello
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
- Department of Orofacial Pain and Dysfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Rodolfo Sardone
- Unit of Statistics and Epidemiology, Local Health Authority of Taranto, Taranto, Italy
| | - Fabio Castellana
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Roberta Zupo
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | | | - Ilaria Bortone
- Local Healthcare Authority of Bari, ASL Bari, Bari, Italy
| | - Roberta Stallone
- Neuroscience and Education, Human Resources Excellence in Research, University of Foggia, Foggia, Italy
| | - Mario Altamura
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Antonello Bellomo
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Antonio Daniele
- Department of Neuroscience, Catholic University of Sacred Heart, Rome, Italy
- Neurology Unit, IRCCS Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Vincenzo Solfrizzi
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Francesco Panza
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
34
|
Molina‐Henry DP, Raman R, Liu A, Langford O, Johnson K, Shum LK, Glover CM, Dhadda S, Irizarry M, Jimenez‐Maggiora G, Braunstein JB, Yarasheski K, Venkatesh V, West T, Verghese PB, Rissman RA, Aisen P, Grill JD, Sperling RA. Racial and ethnic differences in plasma biomarker eligibility for a preclinical Alzheimer's disease trial. Alzheimers Dement 2024; 20:3827-3838. [PMID: 38629508 PMCID: PMC11180863 DOI: 10.1002/alz.13803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 06/18/2024]
Abstract
INTRODUCTION In trials of amyloid-lowering drugs for Alzheimer's disease (AD), differential eligibility may contribute to under-inclusion of racial and ethnic underrepresented groups. We examined plasma amyloid beta 42/40 and positron emission tomography (PET) amyloid eligibility for the ongoing AHEAD Study preclinical AD program (NCT04468659). METHODS Univariate logistic regression models were used to examine group differences in plasma and PET amyloid screening eligibility. RESULTS Of 4905 participants screened at time of analysis, 1724 were plasma eligible to continue in screening: 13.3% Hispanic Black, 24.7% Hispanic White, 20.8% non-Hispanic (NH) Asian, 24.7% NH Black, and 38.9% NH White. Plasma eligibility differed across groups in models controlling for covariates (odds ratio from 1.9 to 4.0 compared to the NH White reference group, P < 0.001). Among plasma eligible participants, PET eligibility did not differ by group. DISCUSSION These results suggest that prevalence of brain amyloid pathology differed, but that eligibility based on plasma was equally effective across racial and ethnic group members. HIGHLIGHTS Plasma amyloid eligibility is lower in underrepresented racial and ethnic groups. In plasma eligible adults, positron emission tomography eligibility rates are similar across race and ethnicity. Plasma biomarker tests may be similarly effective across racial and ethnic groups.
Collapse
Affiliation(s)
- Doris Patricia Molina‐Henry
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
- Present address:
Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern California, 9860 Mesa Rim Rd, San Diego, CA, 92121
| | - Rema Raman
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Andy Liu
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Oliver Langford
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Keith Johnson
- Massachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Brigham and Women's HospitalBostonMassachusettsUSA
| | - Leona K. Shum
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Crystal M. Glover
- Rush Alzheimer's Disease CenterChicagoIllinoisUSA
- Department of Psychiatry and Behavioral SciencesRush University Medical CollegeChicagoIllinoisUSA
- Department of Neurological SciencesRush Medical CollegeChicagoIllinoisUSA
| | | | | | - Gustavo Jimenez‐Maggiora
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | | | | | | | - Tim West
- C2N DiagnosticsSt. LouisMissouriUSA
| | | | - Robert A. Rissman
- Department of Physiology and NeuroscienceAlzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Paul Aisen
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Joshua D. Grill
- Institute for Memory Impairments and Neurological DisordersUniversity of California IrvineIrvineCaliforniaUSA
| | - Reisa A. Sperling
- Massachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Brigham and Women's HospitalBostonMassachusettsUSA
| |
Collapse
|
35
|
Ourry V, Binette AP, St-Onge F, Strikwerda-Brown C, Chagnot A, Poirier J, Breitner J, Arenaza-Urquijo EM, Rabin JS, Buckley R, Gonneaud J, Marchant NL, Villeneuve S. How Do Modifiable Risk Factors Affect Alzheimer's Disease Pathology or Mitigate Its Effect on Clinical Symptom Expression? Biol Psychiatry 2024; 95:1006-1019. [PMID: 37689129 DOI: 10.1016/j.biopsych.2023.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 08/11/2023] [Accepted: 09/03/2023] [Indexed: 09/11/2023]
Abstract
Epidemiological studies show that modifiable risk factors account for approximately 40% of the population variability in risk of developing dementia, including sporadic Alzheimer's disease (AD). Recent findings suggest that these factors may also modify disease trajectories of people with autosomal-dominant AD. With positron emission tomography imaging, it is now possible to study the disease many years before its clinical onset. Such studies can provide key knowledge regarding pathways for either the prevention of pathology or the postponement of its clinical expression. The former "resistance pathway" suggests that modifiable risk factors could affect amyloid and tau burden decades before the appearance of cognitive impairment. Alternatively, the resilience pathway suggests that modifiable risk factors may mitigate the symptomatic expression of AD pathology on cognition. These pathways are not mutually exclusive and may appear at different disease stages. Here, in a narrative review, we present neuroimaging evidence that supports both pathways in sporadic AD and autosomal-dominant AD. We then propose mechanisms for their protective effect. Among possible mechanisms, we examine neural and vascular mechanisms for the resistance pathway. We also describe brain maintenance and functional compensation as bases for the resilience pathway. Improved mechanistic understanding of both pathways may suggest new interventions.
Collapse
Affiliation(s)
- Valentin Ourry
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada.
| | - Alexa Pichet Binette
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada; Clinical Memory Research Unit, Department of Clinical Sciences, Lunds Universitet, Malmö, Sweden
| | - Frédéric St-Onge
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Cherie Strikwerda-Brown
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada; School of Psychological Science, The University of Western Australia, Perth, Western Australia, Australia
| | - Audrey Chagnot
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Judes Poirier
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - John Breitner
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Eider M Arenaza-Urquijo
- Environment and Health over the Lifecourse Programme, Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain; Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Jennifer S Rabin
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada; Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada; Rehabilitation Sciences Institute, University of Toronto, Toronto, Ontario, Canada
| | - Rachel Buckley
- Melbourne School of Psychological Sciences University of Melbourne, Parkville, Victoria, Australia; Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Julie Gonneaud
- Normandie University, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Natalie L Marchant
- Division of Psychiatry, University College London, London, United Kingdom
| | - Sylvia Villeneuve
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada; McConnell Brain Imaging Center, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
36
|
Li Y, Xie L, Khandelwal P, Wisse LEM, Brown CA, Prabhakaran K, Dylan Tisdall M, Mechanic-Hamilton D, Detre JA, Das SR, Wolk DA, Yushkevich PA. Automatic segmentation of medial temporal lobe subregions in multi-scanner, multi-modality MRI of variable quality. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595190. [PMID: 38826413 PMCID: PMC11142184 DOI: 10.1101/2024.05.21.595190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Background Volumetry of subregions in the medial temporal lobe (MTL) computed from automatic segmentation in MRI can track neurodegeneration in Alzheimer's disease. However, image quality may vary in MRI. Poor quality MR images can lead to unreliable segmentation of MTL subregions. Considering that different MRI contrast mechanisms and field strengths (jointly referred to as "modalities" here) offer distinct advantages in imaging different parts of the MTL, we developed a muti-modality segmentation model using both 7 tesla (7T) and 3 tesla (3T) structural MRI to obtain robust segmentation in poor-quality images. Method MRI modalities including 3T T1-weighted, 3T T2-weighted, 7T T1-weighted and 7T T2-weighted (7T-T2w) of 197 participants were collected from a longitudinal aging study at the Penn Alzheimer's Disease Research Center. Among them, 7T-T2w was used as the primary modality, and all other modalities were rigidly registered to the 7T-T2w. A model derived from nnU-Net took these registered modalities as input and outputted subregion segmentation in 7T-T2w space. 7T-T2w images most of which had high quality from 25 selected training participants were manually segmented to train the multi-modality model. Modality augmentation, which randomly replaced certain modalities with Gaussian noise, was applied during training to guide the model to extract information from all modalities. To compare our proposed model with a baseline single-modality model in the full dataset with mixed high/poor image quality, we evaluated the ability of derived volume/thickness measures to discriminate Amyloid+ mild cognitive impairment (A+MCI) and Amyloid- cognitively unimpaired (A-CU) groups, as well as the stability of these measurements in longitudinal data. Results The multi-modality model delivered good performance regardless of 7T-T2w quality, while the single-modality model under-segmented subregions in poor-quality images. The multi-modality model generally demonstrated stronger discrimination of A+MCI versus A-CU. Intra-class correlation and Bland-Altman plots demonstrate that the multi-modality model had higher longitudinal segmentation consistency in all subregions while the single-modality model had low consistency in poor-quality images. Conclusion The multi-modality MRI segmentation model provides an improved biomarker for neurodegeneration in the MTL that is robust to image quality. It also provides a framework for other studies which may benefit from multimodal imaging.
Collapse
Affiliation(s)
- Yue Li
- Penn Image Computing and Science Laboratory, University of Pennsylvania, Philadelphia, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | - Long Xie
- Department of Digital Technology and Innovation, Siemens Healthineers, Princeton, USA
| | - Pulkit Khandelwal
- Penn Image Computing and Science Laboratory, University of Pennsylvania, Philadelphia, USA
| | - Laura E M Wisse
- Department of Diagnostic Radiology, Lund University, Lund, Sweden
| | | | | | - M Dylan Tisdall
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | - Dawn Mechanic-Hamilton
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
- Penn Memory Center, University of Pennsylvania, Philadelphia, USA
| | - John A Detre
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - Sandhitsu R Das
- Penn Image Computing and Science Laboratory, University of Pennsylvania, Philadelphia, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - David A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
- Penn Memory Center, University of Pennsylvania, Philadelphia, USA
| | - Paul A Yushkevich
- Penn Image Computing and Science Laboratory, University of Pennsylvania, Philadelphia, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
37
|
Yu Y, Yu S, Battaglia G, Tian X. Amyloid-β in Alzheimer's disease: Structure, toxicity, distribution, treatment, and prospects. IBRAIN 2024; 10:266-289. [PMID: 39346788 PMCID: PMC11427815 DOI: 10.1002/ibra.12155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 10/01/2024]
Abstract
Amyloid-β (Aβ) is a pivotal biomarker in Alzheimer's disease (AD), attracting considerable attention from numerous researchers. There is uncertainty regarding whether clearing Aβ is beneficial or harmful to cognitive function. This question has been a central topic of research, especially given the lack of success in developing Aβ-targeted drugs for AD. However, with the Food and Drug Administration's approval of Lecanemab as the first anti-Aβ medication in July 2023, there is a significant shift in perspective on the potential of Aβ as a therapeutic target for AD. In light of this advancement, this review aims to illustrate and consolidate the molecular structural attributes and pathological ramifications of Aβ. Furthermore, it elucidates the determinants influencing its expression levels while delineating the gamut of extant Aβ-targeted pharmacotherapies that have been subjected to clinical or preclinical evaluation. Subsequently, a comprehensive analysis is presented, dissecting the research landscape of Aβ across the domains above, culminating in the presentation of informed perspectives. Concluding reflections contemplate the supplementary advantages conferred by nanoparticle constructs, conceptualized within the framework of multivalent theory, within the milieu of AD diagnosis and therapeutic intervention, supplementing conventional modalities.
Collapse
Affiliation(s)
- Yifan Yu
- Institute for Bioengineering of Catalunya (IBEC)The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I ReixacBarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Shilong Yu
- Institute for Bioengineering of Catalunya (IBEC)The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I ReixacBarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Giuseppe Battaglia
- Institute for Bioengineering of Catalunya (IBEC)The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I ReixacBarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Xiaohe Tian
- Institute for Bioengineering of Catalunya (IBEC)The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I ReixacBarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
38
|
Ravichandran S, Snyder PJ, Alber J, Kenny MR, Rothstein A, Brown K, Murchison CF, Clay OJ, Roberson ED, Arthur E. Quantifying Putative Retinal Gliosis in Preclinical Alzheimer's Disease. Invest Ophthalmol Vis Sci 2024; 65:5. [PMID: 38696189 PMCID: PMC11077916 DOI: 10.1167/iovs.65.5.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/09/2024] [Indexed: 05/12/2024] Open
Abstract
Purpose Neuroinflammation plays a significant role in the pathology of Alzheimer's disease (AD). Mouse models of AD and postmortem biopsy of patients with AD reveal retinal glial activation comparable to central nervous system immunoreactivity. We hypothesized that the surface area of putative retinal gliosis observed in vivo using en face optical coherence tomography (OCT) imaging will be larger in patients with preclinical AD versus controls. Methods The Spectralis II instrument was used to acquire macular centered 20 × 20 and 30 × 25-degrees spectral domain OCT images of 76 participants (132 eyes). A cohort of 22 patients with preclinical AD (40 eyes, mean age = 69 years, range = 60-80 years) and 20 control participants (32 eyes, mean age = 66 years, range = 58-82 years, P = 0.11) were included for the assessment of difference in surface area of putative retinal gliosis and retinal nerve fiber layer (RNFL) thickness. The surface area of putative retinal gliosis and RNFL thickness for the nine sectors of the Early Treatment Diabetic Retinopathy Study (ETDRS) map were compared between groups using generalized linear mixed models. Results The surface area of putative retinal gliosis was significantly greater in the preclinical AD group (0.97 ± 0.55 mm2) compared to controls (0.68 ± 0.40 mm2); F(1,70) = 4.41, P = 0.039; Cohen's d = 0.61. There was no significant difference between groups for RNFL thickness in the 9 ETDRS sectors, P > 0.05. Conclusions Our analysis shows greater putative retinal gliosis in preclinical AD compared to controls. This demonstrates putative retinal gliosis as a potential biomarker for AD-related neuroinflammation.
Collapse
Affiliation(s)
- Swetha Ravichandran
- School of Optometry, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Peter J. Snyder
- Department of Neurology, Alpert Medical School of Brown University, Providence, Rhode Island, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, United States
| | - Jessica Alber
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, United States
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island, United States
- Butler Hospital Memory and Aging Program, Providence, Rhode Island, United States
| | - Madelyn R. Kenny
- School of Optometry, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Andrew Rothstein
- School of Optometry, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Keisha Brown
- School of Optometry, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Charles F. Murchison
- Alzheimer's Disease Research Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Olivio J. Clay
- Alzheimer's Disease Research Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Department of Psychology, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Erik D. Roberson
- Alzheimer's Disease Research Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Edmund Arthur
- School of Optometry, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
39
|
Jagust WJ, Mattay VS, Krainak DM, Wang SJ, Weidner LD, Hofling AA, Koo H, Hsieh P, Kuo PH, Farrar G, Marzella L. Quantitative Brain Amyloid PET. J Nucl Med 2024; 65:670-678. [PMID: 38514082 PMCID: PMC11064834 DOI: 10.2967/jnumed.123.265766] [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: 09/19/2023] [Revised: 02/13/2024] [Indexed: 03/23/2024] Open
Abstract
Since the development of amyloid tracers for PET imaging, there has been interest in quantifying amyloid burden in the brains of patients with Alzheimer disease. Quantitative amyloid PET imaging is poised to become a valuable approach in disease staging, theranostics, monitoring, and as an outcome measure for interventional studies. Yet, there are significant challenges and hurdles to overcome before it can be implemented into widespread clinical practice. On November 17, 2022, the U.S. Food and Drug Administration, Society of Nuclear Medicine and Molecular Imaging, and Medical Imaging and Technology Alliance cosponsored a public workshop comprising experts from academia, industry, and government agencies to discuss the role of quantitative brain amyloid PET imaging in staging, prognosis, and longitudinal assessment of Alzheimer disease. The workshop discussed a range of topics, including available radiopharmaceuticals for amyloid imaging; the methodology, metrics, and analytic validity of quantitative amyloid PET imaging; its use in disease staging, prognosis, and monitoring of progression; and challenges facing the field. This report provides a high-level summary of the presentations and the discussion.
Collapse
Affiliation(s)
| | - Venkata S Mattay
- Division of Imaging and Radiation Medicine, Office of Specialty Medicine, Office of New Drugs, Center of Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland;
| | - Daniel M Krainak
- Division of Radiological Imaging and Radiation Therapy Devices, Office of Radiological Health, Office of Product Evaluation and Quality, Centers for Devices and Radiological Health, Food and Drug Administration, Silver Spring, Maryland
| | - Sue-Jane Wang
- Division of Biometrics I, Office of Biostatistics, Office of Translational Sciences, Center of Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Lora D Weidner
- Division of Radiological Imaging and Radiation Therapy Devices, Office of Radiological Health, Office of Product Evaluation and Quality, Centers for Devices and Radiological Health, Food and Drug Administration, Silver Spring, Maryland
| | - A Alex Hofling
- Division of Imaging and Radiation Medicine, Office of Specialty Medicine, Office of New Drugs, Center of Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Hayoung Koo
- Division of Imaging and Radiation Medicine, Office of Specialty Medicine, Office of New Drugs, Center of Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | | | | | | | - Libero Marzella
- Division of Imaging and Radiation Medicine, Office of Specialty Medicine, Office of New Drugs, Center of Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
40
|
2024 Alzheimer's disease facts and figures. Alzheimers Dement 2024; 20:3708-3821. [PMID: 38689398 PMCID: PMC11095490 DOI: 10.1002/alz.13809] [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: 05/02/2024]
Abstract
This article describes the public health impact of Alzheimer's disease (AD), including prevalence and incidence, mortality and morbidity, use and costs of care and the ramifications of AD for family caregivers, the dementia workforce and society. The Special Report discusses the larger health care system for older adults with cognitive issues, focusing on the role of caregivers and non-physician health care professionals. An estimated 6.9 million Americans age 65 and older are living with Alzheimer's dementia today. This number could grow to 13.8 million by 2060, barring the development of medical breakthroughs to prevent or cure AD. Official AD death certificates recorded 119,399 deaths from AD in 2021. In 2020 and 2021, when COVID-19 entered the ranks of the top ten causes of death, Alzheimer's was the seventh-leading cause of death in the United States. Official counts for more recent years are still being compiled. Alzheimer's remains the fifth-leading cause of death among Americans age 65 and older. Between 2000 and 2021, deaths from stroke, heart disease and HIV decreased, whereas reported deaths from AD increased more than 140%. More than 11 million family members and other unpaid caregivers provided an estimated 18.4 billion hours of care to people with Alzheimer's or other dementias in 2023. These figures reflect a decline in the number of caregivers compared with a decade earlier, as well as an increase in the amount of care provided by each remaining caregiver. Unpaid dementia caregiving was valued at $346.6 billion in 2023. Its costs, however, extend to unpaid caregivers' increased risk for emotional distress and negative mental and physical health outcomes. Members of the paid health care and broader community-based workforce are involved in diagnosing, treating and caring for people with dementia. However, the United States faces growing shortages across different segments of the dementia care workforce due to a combination of factors, including the absolute increase in the number of people living with dementia. Therefore, targeted programs and care delivery models will be needed to attract, better train and effectively deploy health care and community-based workers to provide dementia care. Average per-person Medicare payments for services to beneficiaries age 65 and older with AD or other dementias are almost three times as great as payments for beneficiaries without these conditions, and Medicaid payments are more than 22 times as great. Total payments in 2024 for health care, long-term care and hospice services for people age 65 and older with dementia are estimated to be $360 billion. The Special Report investigates how caregivers of older adults with cognitive issues interact with the health care system and examines the role non-physician health care professionals play in facilitating clinical care and access to community-based services and supports. It includes surveys of caregivers and health care workers, focusing on their experiences, challenges, awareness and perceptions of dementia care navigation.
Collapse
|
41
|
Bueichekú E, Diez I, Gagliardi G, Kim CM, Mimmack K, Sepulcre J, Vannini P. Multi-modal Neuroimaging Phenotyping of Mnemonic Anosognosia in the Aging Brain. COMMUNICATIONS MEDICINE 2024; 4:65. [PMID: 38580832 PMCID: PMC10997795 DOI: 10.1038/s43856-024-00497-9] [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: 03/13/2023] [Accepted: 03/28/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Unawareness is a behavioral condition characterized by a lack of self-awareness of objective memory decline. In the context of Alzheimer's Disease (AD), unawareness may develop in predementia stages and contributes to disease severity and progression. Here, we use in-vivo multi-modal neuroimaging to profile the brain phenotype of individuals presenting altered self-awareness of memory during aging. METHODS Amyloid- and tau-PET (N = 335) and resting-state functional MRI (N = 713) imaging data of individuals from the Anti-Amyloid Treatment in Asymptomatic Alzheimer's Disease (A4)/Longitudinal Evaluation of Amyloid Risk and Neurodegeneration (LEARN) Study were used in this research. We applied whole-brain voxel-wise and region-of-interest analyses to characterize the cortical intersections of tau, amyloid, and functional connectivity networks underlying unawareness in the aging brain compared to aware, complainer and control groups. RESULTS Individuals with unawareness present elevated amyloid and tau burden in midline core regions of the default mode network compared to aware, complainer or control individuals. Unawareness is characterized by an altered network connectivity pattern featuring hyperconnectivity in the medial anterior prefrontal cortex and posterior occipito-parietal regions co-locating with amyloid and tau deposition. CONCLUSIONS Unawareness is an early behavioral biomarker of AD pathology. Failure of the self-referential system in unawareness of memory decline can be linked to amyloid and tau burden, along with functional network connectivity disruptions, in several medial frontal and parieto-occipital areas of the human brain.
Collapse
Affiliation(s)
- Elisenda Bueichekú
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ibai Diez
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Geoffroy Gagliardi
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Chan-Mi Kim
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kayden Mimmack
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jorge Sepulcre
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
- Department of Radiology, Yale PET Center, Yale Medical School, Yale University, New Haven, CT, USA.
| | - Patrizia Vannini
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
42
|
Ritchie M, Salazar CR, Gillen DL, Grill JD. Post-disclosure distress among racial and ethnic groups in a preclinical AD trial. Alzheimers Dement 2024; 20:2508-2515. [PMID: 38329007 PMCID: PMC11032552 DOI: 10.1002/alz.13726] [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: 09/01/2023] [Revised: 12/13/2023] [Accepted: 01/06/2024] [Indexed: 02/09/2024]
Abstract
INTRODUCTION Trialists need a thorough understanding of whether reactions to Alzheimer's disease (AD) biomarker information differ among racial and ethnic groups in preclinical AD trials. METHODS We used data from the Anti-Amyloid Treatment in Asymptomatic Alzheimer's Disease Study to analyze cognitively unimpaired participants' responses on the Impact of Event Scale (IES) 24 to 72 hours after amyloid disclosure. We fit a linear regression model to test whether mean IES scores differed among participants from specific racial and ethnic groups. We considered potential effect modification by amyloid status. RESULTS Reactions to disclosure did not significantly differ among participant groups based on self-reported race and ethnicity. Although the results were not significant when stratified by amyloid status, all racial and ethnic groups except for participants self-reporting Hispanic/Latino ethnicity were observed to have higher mean IES in the elevated amyloid group. DISCUSSION These results support continued use of current disclosure methods in preclinical AD trials.
Collapse
Affiliation(s)
- Marina Ritchie
- UC Irvine Institute for Memory Impairments and Neurological DisordersUniversity of California, IrvineIrvineCaliforniaUSA
- Department of Neurobiology and BehaviorUniversity of California, IrvineIrvineCaliforniaUSA
| | - Christian R. Salazar
- UC Irvine Institute for Memory Impairments and Neurological DisordersUniversity of California, IrvineIrvineCaliforniaUSA
| | - Daniel L. Gillen
- UC Irvine Institute for Memory Impairments and Neurological DisordersUniversity of California, IrvineIrvineCaliforniaUSA
- Department of StatisticsUniversity of California, IrvineIrvineCaliforniaUSA
| | - Joshua D. Grill
- UC Irvine Institute for Memory Impairments and Neurological DisordersUniversity of California, IrvineIrvineCaliforniaUSA
- Department of Neurobiology and BehaviorUniversity of California, IrvineIrvineCaliforniaUSA
- Department of Psychiatry and Human BehaviorUniversity of California, IrvineIrvineCaliforniaUSA
| |
Collapse
|
43
|
Mohs RC, Beauregard D, Dwyer J, Gaudioso J, Bork J, MaGee‐Rodgers T, Key MN, Kerwin DR, Hughes L, Cordell CB. The Bio-Hermes Study: Biomarker database developed to investigate blood-based and digital biomarkers in community-based, diverse populations clinically screened for Alzheimer's disease. Alzheimers Dement 2024; 20:2752-2765. [PMID: 38415908 PMCID: PMC11032569 DOI: 10.1002/alz.13722] [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: 10/09/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 02/29/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) trial participants are often screened for eligibility by brain amyloid positron emission tomography/cerebrospinal fluid (PET/CSF), which is inefficient as many are not amyloid positive. Use of blood-based biomarkers may reduce screen failures. METHODS We recruited 755 non-Hispanic White, 115 Hispanic, 112 non-Hispanic Black, and 19 other minority participants across groups of cognitively normal (n = 417), mild cognitive impairment (n = 312), or mild AD (n = 272) participants. Plasma amyloid beta (Aβ)40, Aβ42, Aβ42/Aβ40, total tau, phosphorylated tau (p-tau)181, and p-tau217 were measured; amyloid PET/CSF (n = 956) determined amyloid positivity. Clinical, blood biomarker, and ethnicity/race differences associated with amyloid status were evaluated. RESULTS Greater impairment, older age, and carrying an apolipoprotein E (apoE) ε4 allele were associated with greater amyloid burden. Areas under the receiver operating characteristic curve for amyloid status of plasma Aβ42/Aβ40, p-tau181, and p-tau217 with amyloid positivity were ≥ 0.7117 for all ethnoracial groups (p-tau217, ≥0.8128). Age and apoE ε4 adjustments and imputation of biomarker values outside limit of quantitation provided small improvement in predictive power. DISCUSSION Blood-based biomarkers are highly associated with amyloid PET/CSF results in diverse populations enrolled at clinical trial sites. HIGHLIGHTS Amyloid beta (Aβ)42/Aβ40, phosphorylated tau (p-tau)181, and p-tau 217 blood-based biomarkers predicted brain amyloid positivity. P-tau 217 was the strongest predictor of brain amyloid positivity. Biomarkers from diverse ethnic, racial, and clinical cohorts predicted brain amyloid positivity. Community-based populations have similar Alzheimer's disease (AD) biomarker levels as other populations. A prescreen process with blood-based assays may reduce the number of AD trial screen failures.
Collapse
Grants
- Abbvie, Alzheimer's Drug Discovery Foundation (ADDF), Aural Analytics, Biogen, Cognivue, C2N, Gates Ventures, Linus Health, Merck & Co, Quanterix, Retispec, and Roche
Collapse
Affiliation(s)
- Richard C. Mohs
- Global Alzheimer's Platform FoundationWashingtonDistrict of ColumbiaUSA
| | | | - John Dwyer
- Global Alzheimer's Platform FoundationWashingtonDistrict of ColumbiaUSA
| | - Jennifer Gaudioso
- Global Alzheimer's Platform FoundationWashingtonDistrict of ColumbiaUSA
| | - Jason Bork
- Global Alzheimer's Platform FoundationWashingtonDistrict of ColumbiaUSA
| | | | - Mickeal N. Key
- Global Alzheimer's Platform FoundationWashingtonDistrict of ColumbiaUSA
| | | | - Lynn Hughes
- Advisor to the Global Alzheimer's Platform Foundation and IXICO plcLondonUK
| | - Cyndy B. Cordell
- Advisor to the Global Alzheimer's Platform FoundationWashingtonDistrict of ColumbiaUSA
| |
Collapse
|
44
|
Keegan AP, Stough C, Paris D, Luis CA, Abdullah L, Ait-Ghezala G, Chaykin J, Crawford F, Mullan M. Baseline serum brain-derived neurotrophic factor association with future cognition in community-dwelling older adults undergoing annual memory screening. Neurol Res 2024; 46:253-260. [PMID: 38095353 DOI: 10.1080/01616412.2023.2294581] [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: 04/16/2023] [Accepted: 12/09/2023] [Indexed: 01/24/2024]
Abstract
OBJECTIVES It has been shown that peripheral measures of brain-derived neurotrophic factor (BNDF), an important neurotrophin instrumental to the biology of learning, may contribute to predicting cognitive decline. However, the two primary forms of BDNF, mature (mBDNF) and pro (proBDNF), and how they contribute to cognition longitudinally has not been well studied. METHODS Eighty-two older adults (average age 72.2 ± 6.4 years) provided blood samples at two time points separated on average by 4.2 years while participating in an annual memory screening that included the MoCA (Montreal Cognitive Assessment) and GDS (Geriatric Depression Scale). Both mBDNF and proBDNF from serum were quantified at each time point. Whole blood samples were genotyped for APOE and BDNF Val66Met. RESULTS Using logistic regression analysis controlling for age, sex, baseline MoCA score, APOE, and BDNF, higher baseline mBDNF was associated with subjects whose screening score was near maximum or maximum (as defined by MoCA score of 29 or 30) at the second collection visit. APOE was a significant contributing factor; however, BDNF Val66Met was not. Using a similar logistic regression analysis, baseline proBDNF was not found to be associated with future cognition. DISCUSSION This study further supports that mBDNF measured in the serum of older adults may reflect a protective role while proBDNF requires further investigation.
Collapse
Affiliation(s)
| | - Con Stough
- Centre for Human Psychopharmacology, Swinburne University, Melbourne, Australia
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Wilks H, Benzinger TLS, Schindler SE, Cruchaga C, Morris JC, Hassenstab J. Predictors and outcomes of fluctuations in the clinical dementia rating scale. Alzheimers Dement 2024; 20:2080-2088. [PMID: 38224146 PMCID: PMC10984446 DOI: 10.1002/alz.13679] [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: 09/08/2023] [Revised: 11/15/2023] [Accepted: 12/03/2023] [Indexed: 01/16/2024]
Abstract
INTRODUCTION Reversion, or change in cognitive status from impaired to normal, is common in aging and dementia studies, but it remains unclear what factors predict reversion. METHODS We investigated whether reverters, defined as those who revert from a Clinical Dementia Rating® (CDR®) scale score of 0.5 to CDR 0) differed on cognition and biomarkers from unimpaired participants (always CDR 0) and impaired participants (converted to CDR > 0 and had no reversion events). Models evaluated relationships between biomarker status, apolipoprotein E (APOE) ε4 status, and cognition. Additional models described predictors of reversion and predictors of eventual progression to CDR > 0. RESULTS CDR reversion was associated with younger age, better cognition, and negative amyloid biomarker status. Reverters that eventually progressed to CDR > 0 had more visits, were older, and were more likely to have an APOE ε4 allele. DISCUSSION CDR reversion occupies a transitional phase in disease progression between cognitive normality and overt dementia. Reverters may be ideal candidates for secondary prevention Alzheimer's disease (AD) trials. HIGHLIGHTS Reverters had more longitudinal cognitive decline than those who remained cognitively normal. Predictors of reversion: younger age, better cognition, and negative amyloid biomarker status. Reverting from CDR 0.5 to 0 is a risk factor for future conversion to CDR > 0. CDR reversion may be a transitional phase in Alzheimer's Disease progression. CDR reverters may be ideal for Alzheimer's disease secondary prevention trials.
Collapse
Affiliation(s)
- Hannah Wilks
- Department of Psychological & Brain SciencesWashington University in St. LouisSt. LouisMissouriUSA
- Charles F. and Joanne Knight Alzheimer Disease Research CenterDepartment of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Tammie L. S. Benzinger
- Charles F. and Joanne Knight Alzheimer Disease Research CenterDepartment of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
- Department of RadiologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Suzanne E. Schindler
- Charles F. and Joanne Knight Alzheimer Disease Research CenterDepartment of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Carlos Cruchaga
- Charles F. and Joanne Knight Alzheimer Disease Research CenterDepartment of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
- Department of PsychiatryWashington University School of Medicine1 Barnes Jewish Hospital PlazaSt. LouisMissouriUSA
| | - John C. Morris
- Charles F. and Joanne Knight Alzheimer Disease Research CenterDepartment of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Jason Hassenstab
- Department of Psychological & Brain SciencesWashington University in St. LouisSt. LouisMissouriUSA
- Charles F. and Joanne Knight Alzheimer Disease Research CenterDepartment of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| |
Collapse
|
46
|
Shi H, Mirzaei N, Koronyo Y, Davis MR, Robinson E, Braun GM, Jallow O, Rentsendorj A, Ramanujan VK, Fert-Bober J, Kramerov AA, Ljubimov AV, Schneider LS, Tourtellotte WG, Hawes D, Schneider JA, Black KL, Kayed R, Selenica MLB, Lee DC, Fuchs DT, Koronyo-Hamaoui M. Identification of retinal tau oligomers, citrullinated tau, and other tau isoforms in early and advanced AD and relations to disease status. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.13.579999. [PMID: 38405854 PMCID: PMC10888760 DOI: 10.1101/2024.02.13.579999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Importance This study identifies and quantifies diverse pathological tau isoforms in the retina of both early and advanced-stage Alzheimer's disease (AD) and determines their relationship with disease status. Objective A case-control study was conducted to investigate the accumulation of retinal neurofibrillary tangles (NFTs), paired helical filament (PHF)-tau, oligomeric tau (oligo-tau), hyperphosphorylated tau (p-tau), and citrullinated tau (Cit-tau) in relation to the respective brain pathology and cognitive dysfunction in mild cognitively impaired (MCI) and AD dementia patients versus normal cognition (NC) controls. Design setting and participants Eyes and brains from donors diagnosed with AD, MCI (due to AD), and NC were collected (n=75 in total), along with clinical and neuropathological data. Brain and retinal cross-sections-in predefined superior-temporal and inferior-temporal (ST/IT) subregions-were subjected to histopathology analysis or Nanostring GeoMx digital spatial profiling. Main outcomes and measure Retinal burden of NFTs (pretangles and mature tangles), PHF-tau, p-tau, oligo-tau, and Cit-tau was assessed in MCI and AD versus NC retinas. Pairwise correlations revealed associations between retinal and brain parameters and cognitive status. Results Increased retinal NFTs (1.8-fold, p=0.0494), PHF-tau (2.3-fold, p<0.0001), oligo-tau (9.1-fold, p<0.0001), CitR 209 -tau (4.3-fold, p<0.0001), pSer202/Thr205-tau (AT8; 4.1-fold, p<0.0001), and pSer396-tau (2.8-fold, p=0.0015) were detected in AD patients. Retinas from MCI patients showed significant increases in NFTs (2.0-fold, p=0.0444), CitR 209 -tau (3.5-fold, p=0.0201), pSer396-tau (2.6-fold, p=0.0409), and, moreover, oligo-tau (5.8-fold, p=0.0045). Nanostring GeoMx quantification demonstrated upregulated retinal p-tau levels in MCI patients at phosphorylation sites of Ser214 (2.3-fold, p=0.0060), Ser396 (1.8-fold, p=0.0052), Ser404 (2.4-fold, p=0.0018), and Thr231 (3.3-fold, p=0.0028). Strong correlations were found between retinal tau forms to paired-brain pathology and cognitive status: a) retinal oligo-tau vs. Braak stage (r=0.60, P=0.0002), b) retinal PHF-tau vs. ABC average score (r=0.64, P=0.0043), c) retinal pSer396-tau vs. brain NFTs (r=0.68, P<0.0001), and d) retinal pSer202/Thr205-tau vs. MMSE scores (r= -0.77, P=0.0089). Conclusions and Relevance This study reveals increases in immature and mature retinal tau isoforms in MCI and AD patients, highlighting their relationship with brain pathology and cognition. The data provide strong incentive to further explore retinal tauopathy markers that may be useful for early detection and monitoring of AD staging through noninvasive retinal imaging.
Collapse
|
47
|
Evans SA, Paitel ER, Bhasin R, Nielson KA. Genetic Risk for Alzheimer's Disease Alters Perceived Executive Dysfunction in Cognitively Healthy Middle-Aged and Older Adults. J Alzheimers Dis Rep 2024; 8:267-279. [PMID: 38405345 PMCID: PMC10894609 DOI: 10.3233/adr-230166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/17/2024] [Indexed: 02/27/2024] Open
Abstract
Background Subjective cognitive complaints (SCC) may be an early indicator of future cognitive decline. However, findings comparing SCC and objective cognitive performance have varied, particularly in the memory domain. Even less well established is the relationship between subjective and objective complaints in non-amnestic domains, such as in executive functioning, despite evidence indicating very early changes in these domains. Moreover, particularly early changes in both amnestic and non-amnestic domains are apparent in those carrying the Apolipoprotein-E ɛ4 allele, a primary genetic risk for Alzheimer's disease (AD). Objective This study investigated the role of the ɛ4 allele in the consistency between subjective and objective executive functioning in 54 healthy, cognitively intact, middle-aged and older adults. Methods Participants (Mage = 64.07, SD = 9.27, range = 48-84; ɛ4+ = 18) completed the Frontal Systems Behavior Scale (FrSBe) Executive Dysfunction Scale (EXECDYS) to measure subjective executive functioning (SEF) and multiple executive functioning tasks, which were condensed into a single factor. Results After accounting for age, depression, and anxiety, objective executive functioning performance significantly predicted SEF. Importantly, ɛ4 moderated this effect. Specifically, those carrying the ɛ4 allele had significantly less accurate self-awareness of their executive functioning compared to ɛ4 non-carriers. Conclusions Utilizing an approach that integrates self-evaluation of executive functioning with objective neurocognitive assessment may help identify the earliest signs of impending cognitive decline, particularly in those with genetic risk for AD. Such an approach could sensitively determine those most prone to future cognitive decline prior to symptom onset, when interventions could be most effective.
Collapse
Affiliation(s)
- Sarah A. Evans
- Department of Psychology, Marquette University, Milwaukee, WI, USA
| | | | - Riya Bhasin
- Department of Psychology, Marquette University, Milwaukee, WI, USA
| | | |
Collapse
|
48
|
Thompson E, Schroder A, He T, Shand C, Soskic S, Oxtoby NP, Barkhof F, Alexander DC. Combining multimodal connectivity information improves modelling of pathology spread in Alzheimer's disease. IMAGING NEUROSCIENCE (CAMBRIDGE, MASS.) 2024; 2:1-19. [PMID: 38947941 PMCID: PMC11211996 DOI: 10.1162/imag_a_00089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/07/2023] [Accepted: 01/02/2024] [Indexed: 07/02/2024]
Abstract
Cortical atrophy and aggregates of misfolded tau proteins are key hallmarks of Alzheimer's disease. Computational models that simulate the propagation of pathogens between connected brain regions have been used to elucidate mechanistic information about the spread of these disease biomarkers, such as disease epicentres and spreading rates. However, the connectomes that are used as substrates for these models are known to contain modality-specific false positive and false negative connections, influenced by the biases inherent to the different methods for estimating connections in the brain. In this work, we compare five types of connectomes for modelling both tau and atrophy patterns with the network diffusion model, which are validated against tau PET and structural MRI data from individuals with either mild cognitive impairment or dementia. We then test the hypothesis that a joint connectome, with combined information from different modalities, provides an improved substrate for the model. We find that a combination of multimodal information helps the model to capture observed patterns of tau deposition and atrophy better than any single modality. This is validated with data from independent datasets. Overall, our findings suggest that combining connectivity measures into a single connectome can mitigate some of the biases inherent to each modality and facilitate more accurate models of pathology spread, thus aiding our ability to understand disease mechanisms, and providing insight into the complementary information contained in different measures of brain connectivity.
Collapse
Affiliation(s)
- Elinor Thompson
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| | - Anna Schroder
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| | - Tiantian He
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| | - Cameron Shand
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| | - Sonja Soskic
- UCL Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Neil P. Oxtoby
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| | - Frederik Barkhof
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit, the Netherlands
- UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Daniel C. Alexander
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| | | |
Collapse
|
49
|
Rissman RA, Langford O, Raman R, Donohue MC, Abdel‐Latif S, Meyer MR, Wente‐Roth T, Kirmess KM, Ngolab J, Winston CN, Jimenez‐Maggiora G, Rafii MS, Sachdev P, West T, Yarasheski KE, Braunstein JB, Irizarry M, Johnson KA, Aisen PS, Sperling RA. Plasma Aβ42/Aβ40 and phospho-tau217 concentration ratios increase the accuracy of amyloid PET classification in preclinical Alzheimer's disease. Alzheimers Dement 2024; 20:1214-1224. [PMID: 37932961 PMCID: PMC10916957 DOI: 10.1002/alz.13542] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/15/2023] [Accepted: 09/28/2023] [Indexed: 11/08/2023]
Abstract
INTRODUCTION Incorporating blood-based Alzheimer's disease biomarkers such as tau and amyloid beta (Aβ) into screening algorithms may improve screening efficiency. METHODS Plasma Aβ, phosphorylated tau (p-tau)181, and p-tau217 concentration levels from AHEAD 3-45 study participants were measured using mass spectrometry. Tau concentration ratios for each proteoform were calculated to normalize for inter-individual differences. Receiver operating characteristic (ROC) curve analysis was performed for each biomarker against amyloid positivity, defined by > 20 Centiloids. Mixture of experts analysis assessed the value of including tau concentration ratios into the existing predictive algorithm for amyloid positron emission tomography status. RESULTS The area under the receiver operating curve (AUC) was 0.87 for Aβ42/Aβ40, 0.74 for phosphorylated variant p-tau181 ratio (p-tau181/np-tau181), and 0.92 for phosphorylated variant p-tau217 ratio (p-tau217/np-tau217). The Plasma Predicted Centiloid (PPC), a predictive model including p-tau217/np-tau217, Aβ42/Aβ40, age, and apolipoprotein E improved AUC to 0.95. DISCUSSION Including plasma p-tau217/np-tau217 along with Aβ42/Aβ40 in predictive algorithms may streamline screening preclinical individuals into anti-amyloid clinical trials. CLINICALTRIALS gov Identifier: NCT04468659 HIGHLIGHTS: The addition of plasma phosphorylated variant p-tau217 ratio (p-tau217/np-tau217) significantly improved plasma biomarker algorithms for identifying preclinical amyloid positron emission tomography positivity. Prediction performance at higher NAV Centiloid levels was improved with p-tau217/np-tau217. All models generated for this study are incorporated into the Plasma Predicted Centiloid (PPC) app for public use.
Collapse
Affiliation(s)
- Robert A. Rissman
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
- VA San Diego Healthcare SystemSan DiegoCaliforniaUSA
| | - Oliver Langford
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Rema Raman
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Michael C. Donohue
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Sara Abdel‐Latif
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | | | | | | | - Jennifer Ngolab
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Charisse N. Winston
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Gustavo Jimenez‐Maggiora
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Michael S. Rafii
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | | | - Tim West
- C2N DiagnosticsSt. LouisMissouriUSA
| | | | | | | | - Keith A. Johnson
- Brigham and Women's Hospital, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Paul S. Aisen
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Reisa A. Sperling
- Brigham and Women's Hospital, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | | |
Collapse
|
50
|
Young AL, Oxtoby NP, Garbarino S, Fox NC, Barkhof F, Schott JM, Alexander DC. Data-driven modelling of neurodegenerative disease progression: thinking outside the black box. Nat Rev Neurosci 2024; 25:111-130. [PMID: 38191721 DOI: 10.1038/s41583-023-00779-6] [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: 11/30/2023] [Indexed: 01/10/2024]
Abstract
Data-driven disease progression models are an emerging set of computational tools that reconstruct disease timelines for long-term chronic diseases, providing unique insights into disease processes and their underlying mechanisms. Such methods combine a priori human knowledge and assumptions with large-scale data processing and parameter estimation to infer long-term disease trajectories from short-term data. In contrast to 'black box' machine learning tools, data-driven disease progression models typically require fewer data and are inherently interpretable, thereby aiding disease understanding in addition to enabling classification, prediction and stratification. In this Review, we place the current landscape of data-driven disease progression models in a general framework and discuss their enhanced utility for constructing a disease timeline compared with wider machine learning tools that construct static disease profiles. We review the insights they have enabled across multiple neurodegenerative diseases, notably Alzheimer disease, for applications such as determining temporal trajectories of disease biomarkers, testing hypotheses about disease mechanisms and uncovering disease subtypes. We outline key areas for technological development and translation to a broader range of neuroscience and non-neuroscience applications. Finally, we discuss potential pathways and barriers to integrating disease progression models into clinical practice and trial settings.
Collapse
Affiliation(s)
- Alexandra L Young
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK.
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Neil P Oxtoby
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK.
| | - Sara Garbarino
- Life Science Computational Laboratory, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Frederik Barkhof
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Jonathan M Schott
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Daniel C Alexander
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| |
Collapse
|