1
|
Chun MY, Park YH, Kim HJ, Na DL, Kim JP, Seo SW, Jang H. Distinct Characteristics of Suspected Non-Alzheimer Pathophysiology in Relation to Cognitive Status and Cerebrovascular Burden. Clin Nucl Med 2025; 50:368-380. [PMID: 40025666 PMCID: PMC11969373 DOI: 10.1097/rlu.0000000000005793] [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: 11/18/2024] [Accepted: 01/23/2025] [Indexed: 03/04/2025]
Abstract
PURPOSE OF THE REPORT This study investigated the prevalence and clinical characteristics of suspected non-Alzheimer disease pathophysiology (SNAP) across varying cognitive statuses and cerebral small vessel disease (CSVD) burden. PATIENTS AND METHODS We included 1992 participants with cognitive status categorized as cognitively unimpaired, mild cognitive impairment, or dementia. β-amyloid (Aβ, A) positivity was assessed by Aβ PET, and neurodegeneration (N) positivity was determined through hippocampal volume. Participants were further divided by the presence or absence of severe CSVD. The clinical and imaging characteristics of A-N+ (SNAP) group were compared with those of the A-N- and A+N+ groups. RESULTS SNAP participants were older and had more vascular risk factors compared with A-N- and A+N+ in the CSVD(-) cohort. SNAP and A+N+ showed similar cortical thinning. At the dementia stage, SNAP had a cognitive trajectory similar to A+N+ in the CSVD(-) cohort. However, SNAP exhibited less cognitive decline than A+N+ in the CSVD(+) cohort. CONCLUSIONS SNAP is characterized by distinct clinical and imaging characteristics; however, it does not necessarily indicate a benign prognosis, particularly at the dementia stage. These findings highlight the need to assess SNAP in relation to the cognitive stage and CSVD presence to better understand its progression and guide interventions.
Collapse
Affiliation(s)
- Min Young Chun
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine
- Department of Neurology, Yonsei University College of Medicine
| | - Yu Hyun Park
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Neuroscience Center, Samsung Medical Center
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University
- Department of Digital Health, SAIHST, Sungkyunkwan University
| | - Duk L. Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
| | - Jun Pyo Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University
- Department of Digital Health, SAIHST, Sungkyunkwan University
- Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Gangnam-gu
| | - Hyemin Jang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Jongno-gu, Seoul, South Korea
| |
Collapse
|
2
|
Lim YY, Mills A, Norfolk M, Rosenich E, Maruff P. Factors influencing rates of unsupervised assessment of short-term learning in cognitively unimpaired adults. J Alzheimers Dis 2025; 103:441-451. [PMID: 39686610 DOI: 10.1177/13872877241302491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
BACKGROUND In older adults with preclinical Alzheimer's disease (AD), learning curves derived from validated psychological learning paradigms are reduced to an extent greater than impairment, or decline, on neuropsychological memory tests. OBJECTIVE This study aimed to examine how age, sex, education, mood, and general dementia risk, which also increases risk for preclinical AD, could influence learning curves. METHODS 1050 adults enrolled in the BetterBrains trial completed 10 blocks of ORCA-LLT learning trials over 5 days. Learning curves were derived from improvement in accuracy over trials. Participants also completed questionnaires of demography and mood, and the CAIDE risk score was computed for each participant. RESULTS Most participants (67%) completed ≥6 blocks of ORCA-LLT. Older age (d = 0.75), lower education (d = 0.50), and higher dementia risk (d = 0.36) were associated significantly with slower learning rates. CONCLUSIONS In older adults, learning curves are influenced subtly by age, education, and dementia risk but not by sex or mood.
Collapse
Affiliation(s)
- Yen Ying Lim
- School of Psychological Sciences, Monash University, Clayton, Australia
- Turner Institute for Brain and Mental Health, Monash University, Clayton, Australia
- ARC Centre for Optimal Ageing, Monash University, Clayton, Australia
| | - Andrea Mills
- School of Psychological Sciences, Monash University, Clayton, Australia
- Department of Psychological Sciences, Swinburne University of Technology, Hawthorn, Australia
| | - Maya Norfolk
- School of Psychological Sciences, Monash University, Clayton, Australia
- Turner Institute for Brain and Mental Health, Monash University, Clayton, Australia
| | - Emily Rosenich
- School of Psychological Sciences, Monash University, Clayton, Australia
- Turner Institute for Brain and Mental Health, Monash University, Clayton, Australia
- ARC Centre for Optimal Ageing, Monash University, Clayton, Australia
| | - Paul Maruff
- Turner Institute for Brain and Mental Health, Monash University, Clayton, Australia
- ARC Centre for Optimal Ageing, Monash University, Clayton, Australia
- Cogstate Ltd, Melbourne, Australia
| |
Collapse
|
3
|
Keith CM, Haut MW, Vieira Ligo Teixeira C, Mehta RI, Phelps H, Ward M, Miller M, Navia RO, Coleman MM, Marano G, Wang X, Pockl S, Rajabalee N, Scarisbrick DM, McCuddy WT, D'Haese PF, Rezai A, Wilhelmsen K. Memory consolidation, temporal and parietal atrophy, and metabolism in amyloid-β positive and negative mild cognitive impairment. J Alzheimers Dis 2024; 102:778-791. [PMID: 39670736 DOI: 10.1177/13872877241291223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is classically characterized by alterations in memory consolidation. With the advent of diagnostic biomarkers, some patients clinically diagnosed with AD display biomarkers inconsistent with the diagnosis. OBJECTIVE We aimed to explore differences in memory consolidation and neurodegeneration of the temporal and parietal lobes as a function of amyloid-β status in amnestic mild cognitive impairment (aMCI). METHODS We examined differences in memory consolidation and neurodegeneration between patients diagnosed with amyloid-β positive aMCI (Aβ+ N = 78), amyloid-β negative aMCI (Aβ- N = 48), and healthy participants (HP; N = 41), within a well-characterized clinical cohort. RESULTS Aβ+ exhibited more pronounced consolidation impairments compared to Aβ-, while Aβ- faced more consolidation challenges than HP. Both Aβ+ and Aβ- were similar in hippocampal volume and entorhinal thickness, but Aβ+ had thinner inferior parietal cortex than Aβ-. Using 18F-fluoro-deoxyglucose-positron emission tomography, metabolism in both temporal and parietal regions was lower in Aβ+ relative to Aβ-. CONCLUSIONS These findings suggest pathologies other than AD likely contribute to memory consolidation difficulties in aMCI, and neurodegeneration of the parietal cortex in combination with hypometabolism may contribute to more pronounced consolidation problems in Aβ+.
Collapse
Affiliation(s)
- Cierra M Keith
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Behavioral Medicine and Psychiatry, West Virginia University, Morgantown, WV, USA
| | - Marc W Haut
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Behavioral Medicine and Psychiatry, West Virginia University, Morgantown, WV, USA
- Department of Neurology, West Virginia University, Morgantown, WV, USA
| | | | - Rashi I Mehta
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Neuroradiology, West Virginia University, Morgantown, WV, USA
| | - Holly Phelps
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Behavioral Medicine and Psychiatry, West Virginia University, Morgantown, WV, USA
| | - Melanie Ward
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Neurology, West Virginia University, Morgantown, WV, USA
| | - Mark Miller
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Behavioral Medicine and Psychiatry, West Virginia University, Morgantown, WV, USA
| | - R Osvaldo Navia
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Medicine, West Virginia University, Morgantown, WV, USA
| | - Michelle M Coleman
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| | - Gary Marano
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Neuroradiology, West Virginia University, Morgantown, WV, USA
- Department of Radiology, West Virginia University, Morgantown, WV, USA
| | - Xiaofei Wang
- Department of Radiology, West Virginia University, Morgantown, WV, USA
| | - Stephanie Pockl
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Medicine, West Virginia University, Morgantown, WV, USA
| | - Nafiisah Rajabalee
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Medicine, West Virginia University, Morgantown, WV, USA
| | - David M Scarisbrick
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Behavioral Medicine and Psychiatry, West Virginia University, Morgantown, WV, USA
| | - William T McCuddy
- Department of Neuropsychology, Barrow Neurological Institute, St Joseph Hospital and Medical Center, Phoenix, AZ, USA
| | - Pierre-François D'Haese
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Neuroradiology, West Virginia University, Morgantown, WV, USA
| | - Ali Rezai
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Neurosurgery, West Virginia University, Morgantown, WV, USA
| | - Kirk Wilhelmsen
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Neurology, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
4
|
Shirzadi Z, Boyle R, Yau WYW, Coughlan G, Fu JF, Properzi MJ, Buckley RF, Yang HS, Scanlon CE, Hsieh S, Amariglio RE, Papp K, Rentz D, Price JC, Johnson KA, Sperling RA, Chhatwal JP, Schultz AP. Vascular contributions to cognitive decline: Beyond amyloid and tau in the Harvard Aging Brain Study. J Cereb Blood Flow Metab 2024; 44:1319-1328. [PMID: 38452039 PMCID: PMC11342726 DOI: 10.1177/0271678x241237624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 03/09/2024]
Abstract
In addition to amyloid and tau pathology, elevated systemic vascular risk, white matter injury, and reduced cerebral blood flow contribute to late-life cognitive decline. Given the strong collinearity among these parameters, we proposed a framework to extract the independent latent features underlying cognitive decline using the Harvard Aging Brain Study (N = 166 cognitively unimpaired older adults at baseline). We used the following measures from the baseline visit: cortical amyloid, inferior temporal cortex tau, relative cerebral blood flow, white matter hyperintensities, peak width of skeletonized mean diffusivity, and Framingham Heart Study cardiovascular disease risk. We used exploratory factor analysis to extract orthogonal factors from these variables and their interactions. These factors were used in a regression model to explain longitudinal Preclinical Alzheimer Cognitive Composite-5 (PACC) decline (follow-up = 8.5 ±2.7 years). We next examined whether gray matter volume atrophy acts as a mediator of factors and PACC decline. Latent factors of systemic vascular risk, white matter injury, and relative cerebral blood flow independently explain cognitive decline beyond amyloid and tau. Gray matter volume atrophy mediates these associations with the strongest effect on white matter injury. These results suggest that systemic vascular risk contributes to cognitive decline beyond current markers of cerebrovascular injury, amyloid, and tau.
Collapse
Affiliation(s)
- Zahra Shirzadi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rory Boyle
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Wai-Ying W Yau
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Gillian Coughlan
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jessie Fanglu Fu
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael J Properzi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rachel F Buckley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hyun-Sik Yang
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Catherine E Scanlon
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephanie Hsieh
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rebecca E Amariglio
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathryn Papp
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Dorene Rentz
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Julie C Price
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Keith A Johnson
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Xia Y, Dore V, Fripp J, Bourgeat P, Laws SM, Fowler CJ, Rainey-Smith SR, Martins RN, Rowe C, Masters CL, Coulson EJ, Maruff P. Association of Basal Forebrain Atrophy With Cognitive Decline in Early Alzheimer Disease. Neurology 2024; 103:e209626. [PMID: 38885444 PMCID: PMC11254448 DOI: 10.1212/wnl.0000000000209626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/09/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND AND OBJECTIVES In early Alzheimer disease (AD), β-amyloid (Aβ) deposition is associated with volume loss in the basal forebrain (BF) and cognitive decline. However, the extent to which Aβ-related BF atrophy manifests as cognitive decline is not understood. This study sought to characterize the relationship between BF atrophy and the decline in memory and attention in patients with early AD. METHODS Participants from the Australian Imaging, Biomarkers and Lifestyle (AIBL) study who completed Aβ-PET imaging and repeated MRI and cognitive assessments were included. At baseline, participants were classified based on their clinical dementia stage and Aβ status, yielding groups that were cognitively unimpaired (CU) Aβ-, CU Aβ+, and mild cognitive impairment (MCI) Aβ+. Linear mixed-effects models were used to assess changes in volumetric measures of BF subregions and the hippocampus and changes in AIBL memory and attention composite scores for each group compared with CU Aβ- participants. Associations between Aβ burden, brain atrophy, and cognitive decline were evaluated and explored further using mediation analyses. RESULTS The cohort included 476 participants (72.6 ± 5.9 years, 55.0% female) with longitudinal data from a median follow-up period of 6.1 years. Compared with the CU Aβ- group (n = 308), both CU Aβ+ (n = 107) and MCI Aβ+ (n = 61) adults showed faster decline in BF and hippocampal volumes and in memory and attention (Cohen d = 0.73-1.74). Rates of atrophy in BF subregions and the hippocampus correlated with cognitive decline, and each individually mediated the impact of Aβ burden on memory and attention decline. When all mediators were considered simultaneously, hippocampal atrophy primarily influenced the effect of Aβ burden on memory decline (β [SE] = -0.139 [0.032], proportion mediated [PM] = 28.0%) while the atrophy of the posterior nucleus basalis of Meynert in the BF (β [SE] = -0.068 [0.029], PM = 13.1%) and hippocampus (β [SE] = -0.121 [0.033], PM = 23.4%) distinctively influenced Aβ-related attention decline. DISCUSSION These findings highlight the significant role of BF atrophy in the complex pathway linking Aβ to cognitive impairment in early stages of AD. Volumetric assessment of BF subregions could be essential in elucidating the relationships between the brain structure and behavior in AD.
Collapse
Affiliation(s)
- Ying Xia
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Vincent Dore
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Jurgen Fripp
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Pierrick Bourgeat
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Simon M Laws
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Christopher J Fowler
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Stephanie R Rainey-Smith
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Ralph N Martins
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Christopher Rowe
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Colin L Masters
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Elizabeth J Coulson
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Paul Maruff
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| |
Collapse
|
6
|
Vos SJB, Delvenne A, Jack CR, Thal DR, Visser PJ. The clinical importance of suspected non-Alzheimer disease pathophysiology. Nat Rev Neurol 2024; 20:337-346. [PMID: 38724589 DOI: 10.1038/s41582-024-00962-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 06/06/2024]
Abstract
The development of biomarkers for Alzheimer disease (AD) has led to the origin of suspected non-AD pathophysiology (SNAP) - a heterogeneous biomarker-based concept that describes individuals with normal amyloid and abnormal tau and/or neurodegeneration biomarker status. In this Review, we describe the origins of the SNAP construct, along with its prevalence, diagnostic and prognostic implications, and underlying neuropathology. As we discuss, SNAP can be operationalized using different biomarker modalities, which could affect prevalence estimates and reported characteristics of SNAP in ways that are not yet fully understood. Moreover, the underlying aetiologies that lead to a SNAP biomarker profile, and whether SNAP is the same in people with and without cognitive impairment, remains unclear. Improved insight into the clinical characteristics and pathophysiology of SNAP is of major importance for research and clinical practice, as well as for trial design to optimize care and treatment of individuals with SNAP.
Collapse
Affiliation(s)
- Stephanie J B Vos
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands.
| | - Aurore Delvenne
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Dietmar R Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology and Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Pathology, University Hospital Leuven, Leuven, Belgium
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
| |
Collapse
|
7
|
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
|
8
|
Kim HH, Kwon MJ, Jo S, Park JE, Kim JW, Kim JH, Kim SE, Kim KW, Han JW. Exploration of neuroanatomical characteristics to differentiate prodromal Alzheimer's disease from cognitively unimpaired amyloid-positive individuals. Sci Rep 2024; 14:10083. [PMID: 38698190 PMCID: PMC11066072 DOI: 10.1038/s41598-024-60843-8] [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: 11/28/2023] [Accepted: 04/28/2024] [Indexed: 05/05/2024] Open
Abstract
Differentiating clinical stages based solely on positive findings from amyloid PET is challenging. We aimed to investigate the neuroanatomical characteristics at the whole-brain level that differentiate prodromal Alzheimer's disease (AD) from cognitively unimpaired amyloid-positive individuals (CU A+) in relation to amyloid deposition and regional atrophy. We included 45 CU A+ participants and 135 participants with amyloid-positive prodromal AD matched 1:3 by age, sex, and education. All participants underwent 18F-florbetaben positron emission tomography and 3D structural T1-weighted magnetic resonance imaging. We compared the standardized uptake value ratios (SUVRs) and volumes in 80 regions of interest (ROIs) between CU A+ and prodromal AD groups using independent t-tests, and employed the least absolute selection and shrinkage operator (LASSO) logistic regression model to identify ROIs associated with prodromal AD in relation to amyloid deposition, regional atrophy, and their interaction. After applying False Discovery Rate correction at < 0.1, there were no differences in global and regional SUVR between CU A+ and prodromal AD groups. Regional volume differences between the two groups were observed in the amygdala, hippocampus, entorhinal cortex, insula, parahippocampal gyrus, and inferior temporal and parietal cortices. LASSO logistic regression model showed significant associations between prodromal AD and atrophy in the entorhinal cortex, inferior parietal cortex, both amygdalae, and left hippocampus. The mean SUVR in the right superior parietal cortex (beta coefficient = 0.0172) and its interaction with the regional volume (0.0672) were also selected in the LASSO model. The mean SUVR in the right superior parietal cortex was associated with an increased likelihood of prodromal AD (Odds ratio [OR] 1.602, p = 0.014), particularly in participants with lower regional volume (OR 3.389, p < 0.001). Only regional volume differences, not amyloid deposition, were observed between CU A+ and prodromal AD. The reduced volume in the superior parietal cortex may play a significant role in the progression to prodromal AD through its interaction with amyloid deposition in that region.
Collapse
Affiliation(s)
- Hak Hyeon Kim
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, 82 Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
- Department of Psychiatry, College of Medicine, Seoul National University, Seoul, South Korea
| | - Min Jeong Kwon
- Department of Brain and Cognitive Science, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Sungman Jo
- Department of Health Science and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Ji Eun Park
- Department of Brain and Cognitive Science, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Ji Won Kim
- Department of Brain and Cognitive Science, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Jae Hyoung Kim
- Department of Radiology, Seoul National University Bundang Hospital, College of Medicine, Seoul National University, Seongnam-si, Gyeonggi-do, South Korea
| | - Sang Eun Kim
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, College of Medicine, Seoul National University, Seongnam-si, Gyeonggi-do, Korea
- Center for Nanomolecular Imaging and Innovative Drug Development, Advanced Institutes of Convergence Technology, Suwon, Republic of Korea
| | - Ki Woong Kim
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, 82 Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
- Department of Psychiatry, College of Medicine, Seoul National University, Seoul, South Korea
- Department of Brain and Cognitive Science, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Department of Health Science and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Ji Won Han
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, 82 Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea.
- Department of Psychiatry, College of Medicine, Seoul National University, Seoul, South Korea.
| |
Collapse
|
9
|
Li JQ, Song JH, Suckling J, Wang YJ, Zuo CT, Zhang C, Gao J, Song YQ, Xie AM, Tan L, Yu JT. Disease trajectories in older adults with non-AD pathologic change and comparison with Alzheimer's disease pathophysiology: A longitudinal study. Neurobiol Aging 2024; 134:106-114. [PMID: 38056216 DOI: 10.1016/j.neurobiolaging.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 12/08/2023]
Abstract
Based on the 'AT(N)' system, individuals with normal amyloid biomarkers but abnormal tauopathy or neurodegeneration biomarkers are classified as non-Alzheimer's disease (AD) pathologic change. This study aimed to assess the long-term clinical and cognitive trajectories of individuals with non-AD pathologic change among older adults without dementia, comparing them to those with normal AD biomarkers and AD pathophysiology. Analyzing Alzheimer's Disease Neuroimaging Initiative data, we evaluated clinical outcomes and conversion risk longitudinally using mixed effects models and multivariate Cox proportional hazard models. We found that compared to individuals with A-T-N-, those with abnormal tauopathy or neurodegeneration biomarkers (A-T + N-, A-T-N + , and A-T + N + ) had a faster rate of cognitive decline and disease progression. Individuals with A-T + N + had a faster rate of decline than those with A-T + N-. Additionally, in individuals with the same baseline tauopathy and neurodegeneration biomarker status, the presence of baseline amyloid could accelerate cognitive decline and clinical progression. These findings provide a foundation for future studies on non-AD pathologic change and its comparison with AD pathophysiology.
Collapse
Affiliation(s)
- Jie-Qiong Li
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China.
| | - Jing-Hui Song
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - John Suckling
- Department of Psychiatry, University of Cambridge, Cambridge CB2 1TN, UK; Medical Research Council and Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge CB2 1TN, UK; Cambridgeshire and Peterborough NHS Trust, UK
| | - Yan-Jiang Wang
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Chuan-Tao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai 200433, China
| | - Can Zhang
- Genetics and Aging Research Unit, Mass GeneralInstitute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown 02138, MA 02129-2060, USA
| | - Jing Gao
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Yu-Qiang Song
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - An-Mu Xie
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital,Qingdao University, Qingdao 266000, Shandong, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for NeurologicalDisorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai 200040, China.
| |
Collapse
|
10
|
Zhang T, Dolga AM, Eisel ULM, Schmidt M. Novel crosstalk mechanisms between GluA3 and Epac2 in synaptic plasticity and memory in Alzheimer's disease. Neurobiol Dis 2024; 191:106389. [PMID: 38142840 DOI: 10.1016/j.nbd.2023.106389] [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: 11/23/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease which accounts for the most cases of dementia worldwide. Impaired memory, including acquisition, consolidation, and retrieval, is one of the hallmarks in AD. At the cellular level, dysregulated synaptic plasticity partly due to reduced long-term potentiation (LTP) and enhanced long-term depression (LTD) underlies the memory deficits in AD. GluA3 containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) are one of key receptors involved in rapid neurotransmission and synaptic plasticity. Recent studies revealed a novel form of GluA3 involved in neuronal plasticity that is dependent on cyclic adenosine monophosphate (cAMP), rather than N-methyl-d-aspartate (NMDA). However, this cAMP-dependent GluA3 pathway is specifically and significantly impaired by amyloid beta (Aβ), a pathological marker of AD. cAMP is a key second messenger that plays an important role in modulating memory and synaptic plasticity. We previously reported that exchange protein directly activated by cAMP 2 (Epac2), acting as a main cAMP effector, plays a specific and time-limited role in memory retrieval. From electrophysiological perspective, Epac2 facilities the maintenance of LTP, a cellular event closely associated with memory retrieval. Additionally, Epac2 was found to be involved in the GluA3-mediated plasticity. In this review, we comprehensively summarize current knowledge regarding the specific roles of GluA3 and Epac2 in synaptic plasticity and memory, and their potential association with AD.
Collapse
Affiliation(s)
- Tong Zhang
- Department of Molecular Pharmacology, University of Groningen, the Netherlands; Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen 9747 AG, Netherlands
| | - Amalia M Dolga
- Department of Molecular Pharmacology, University of Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen 9747 AG, Netherlands
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
11
|
Yuyama K, Sun H, Fujii R, Hemmi I, Ueda K, Igeta Y. Extracellular vesicle proteome unveils cathepsin B connection to Alzheimer's disease pathogenesis. Brain 2024; 147:627-636. [PMID: 38071653 PMCID: PMC10834236 DOI: 10.1093/brain/awad361] [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: 04/26/2023] [Revised: 09/27/2023] [Accepted: 10/10/2023] [Indexed: 02/03/2024] Open
Abstract
Extracellular vesicles (EVs) are membrane vesicles that are released extracellularly and considered to be implicated in the pathogenesis of neurodegenerative diseases including Alzheimer's disease. Here, CSF EVs of 16 ATN-classified cases were subjected to quantitative proteome analysis. In these CSF EVs, levels of 11 proteins were significantly altered during the ATN stage transitions (P < 0.05 and fold-change > 2.0). These proteins were thought to be associated with Alzheimer's disease pathogenesis and represent candidate biomarkers for pathogenic stage classification. Enzyme-linked immunosorbent assay analysis of CSF and plasma EVs revealed altered levels of cathepsin B (CatB) during the ATN transition (seven ATN groups in validation set, n = 136). The CSF and plasma EV CatB levels showed a negative correlation with CSF amyloid-β42 concentrations. This proteomic landscape of CSF EVs in ATN classifications can depict the molecular framework of Alzheimer's disease progression, and CatB may be considered a promising candidate biomarker and therapeutic target in Alzheimer's disease amyloid pathology.
Collapse
Affiliation(s)
- Kohei Yuyama
- Lipid Biofunction Section, Faculty of Advanced Life Science, Hokkaido University, Sapporo 001-0021, Japan
| | - Hui Sun
- Lipid Biofunction Section, Faculty of Advanced Life Science, Hokkaido University, Sapporo 001-0021, Japan
| | - Risa Fujii
- Cancer Proteomics Group, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo 035-8550, Japan
| | - Isao Hemmi
- Department of Nursing, Japanese Red Cross College of Nursing, Tokyo 150-0012, Japan
| | - Koji Ueda
- Cancer Proteomics Group, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo 035-8550, Japan
| | - Yukifusa Igeta
- Department of Dementia, Dementia Center, Toranomon Hospital, Tokyo 105-8470, Japan
- Division of Dementia Research, Okinaka Memorial Institute for Medical Research, Tokyo 105-8470, Japan
| |
Collapse
|
12
|
Wu KY, Lin KJ, Chen CH, Liu CY, Wu YM, Yen TC, Hsiao IT. Atrophy, hypometabolism and implication regarding pathology in late-life major depression with suspected non-alzheimer pathophysiology (SNAP). Biomed J 2023; 46:100589. [PMID: 36914051 PMCID: PMC10749882 DOI: 10.1016/j.bj.2023.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 07/16/2022] [Accepted: 03/08/2023] [Indexed: 03/13/2023] Open
Abstract
BACKGROUND A substantial proportion of individuals with late-life major depression could be classified as having a suspected non-Alzheimer disease pathophysiology (SNAP), as indicated by a negative test for the biomarker β-amyloid (Aβ-) but a positive test for neurodegeneration (ND+). This study investigated the clinical features, characteristic patterns of brain atrophy and hypometabolism, and implications regarding pathology in this population. METHODS Forty-six amyloid-negative patients with late-life major depressive disorder (MDD) patients, including 23 SNAP (Aβ-/ND+) and 23 Aβ-/ND- MDD subjects, and 22 Aβ-/ND-healthy control subjects were included in this study. Voxel-wise group comparisons between the SNAP MDD, Aβ-/ND- MDD and control subjects were performed, adjusting for age, gender and level of education. For exploratory comparisons, 8 Aβ+/ND- and 4 Aβ+/ND + MDD patients were included in the Supplementary Material. RESULTS The SNAP MDD patients had atrophy extending to regions outside the hippocampus, predominately in the medial temporal, dorsomedial and ventromedial prefrontal cortex; hypometabolism involving a large portion of the lateral and medial prefrontal cortex in addition to the bilateral temporal, parietal and precuneus cortex within typical Alzheimer disease regions were observed. Metabolism ratios of the inferior to the medial temporal lobe were significantly elevated in the SNAP MDD patients. We further discussed the implications with regards to underlying pathologies. CONCLUSION The present study demonstrated characteristic patterns of atrophy and hypometabolism in patients with late-life major depression with SNAP. Identifying individuals with SNAP MDD may provide insights into currently unspecified neurodegenerative processes. Future refinement of neurodegeneration biomarkers is essential in order to identify potential pathological correlates while in vivo reliable pathological biomarkers are not forthcoming.
Collapse
Affiliation(s)
- Kuan-Yi Wu
- Department of Psychiatry, Chang Gung Memorial Hospital, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kun-Ju Lin
- Department of Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Department of Medical Imaging and Radiological Sciences and Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan; Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chia-Hsiang Chen
- Department of Psychiatry, Chang Gung Memorial Hospital, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Yih Liu
- Department of Psychiatry, Chang Gung Memorial Hospital, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Ming Wu
- Department of Radiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Tzu-Chen Yen
- Department of Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Department of Medical Imaging and Radiological Sciences and Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan; APRINOIA Therapeutics Inc., Taipei, Taiwan
| | - Ing-Tsung Hsiao
- Department of Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Department of Medical Imaging and Radiological Sciences and Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
13
|
Gouilly D, Rafiq M, Nogueira L, Salabert AS, Payoux P, Péran P, Pariente J. Beyond the amyloid cascade: An update of Alzheimer's disease pathophysiology. Rev Neurol (Paris) 2023; 179:812-830. [PMID: 36906457 DOI: 10.1016/j.neurol.2022.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 10/02/2022] [Accepted: 12/02/2022] [Indexed: 03/13/2023]
Abstract
Alzheimer's disease (AD) is a multi-etiology disease. The biological system of AD is associated with multidomain genetic, molecular, cellular, and network brain dysfunctions, interacting with central and peripheral immunity. These dysfunctions have been primarily conceptualized according to the assumption that amyloid deposition in the brain, whether from a stochastic or a genetic accident, is the upstream pathological change. However, the arborescence of AD pathological changes suggests that a single amyloid pathway might be too restrictive or inconsistent with a cascading effect. In this review, we discuss the recent human studies of late-onset AD pathophysiology in an attempt to establish a general updated view focusing on the early stages. Several factors highlight heterogenous multi-cellular pathological changes in AD, which seem to work in a self-amplifying manner with amyloid and tau pathologies. Neuroinflammation has an increasing importance as a major pathological driver, and perhaps as a convergent biological basis of aging, genetic, lifestyle and environmental risk factors.
Collapse
Affiliation(s)
- D Gouilly
- Toulouse Neuroimaging Center, Toulouse, France.
| | - M Rafiq
- Toulouse Neuroimaging Center, Toulouse, France; Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France
| | - L Nogueira
- Department of Cell Biology and Cytology, CHU Toulouse Purpan, France
| | - A-S Salabert
- Toulouse Neuroimaging Center, Toulouse, France; Department of Nuclear Medicine, CHU Toulouse Purpan, France
| | - P Payoux
- Toulouse Neuroimaging Center, Toulouse, France; Department of Nuclear Medicine, CHU Toulouse Purpan, France; Center of Clinical Investigation, CHU Toulouse Purpan (CIC1436), France
| | - P Péran
- Toulouse Neuroimaging Center, Toulouse, France
| | - J Pariente
- Toulouse Neuroimaging Center, Toulouse, France; Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France; Center of Clinical Investigation, CHU Toulouse Purpan (CIC1436), France
| |
Collapse
|
14
|
Gagliardi G, Rodriguez-Vieitez E, Montal V, Sepulcre J, Diez I, Lois C, Hanseeuw B, Schultz AP, Properzi MJ, Papp KV, Marshall GA, Fortea J, Johnson KA, Sperling RA, Vannini P. Cortical microstructural changes predict tau accumulation and episodic memory decline in older adults harboring amyloid. COMMUNICATIONS MEDICINE 2023; 3:106. [PMID: 37528163 PMCID: PMC10394044 DOI: 10.1038/s43856-023-00324-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/19/2023] [Indexed: 08/03/2023] Open
Abstract
INTRODUCTION Non-invasive diffusion-weighted imaging (DWI) to assess brain microstructural changes via cortical mean diffusivity (cMD) has been shown to be cross-sectionally associated with tau in cognitively normal older adults, suggesting that it might be an early marker of neuronal injury. Here, we investigated how regional cortical microstructural changes measured by cMD are related to the longitudinal accumulation of regional tau as well as to episodic memory decline in cognitively normal individuals harboring amyloid pathology. METHODS 122 cognitively normal participants from the Harvard Aging Brain Study underwent DWI, T1w-MRI, amyloid and tau PET imaging, and Logical Memory Delayed Recall (LMDR) assessments. We assessed whether the interaction of baseline amyloid status and cMD (in entorhinal and inferior-temporal cortices) was associated with longitudinal regional tau accumulation and with longitudinal LMDR using separate linear mixed-effects models. RESULTS We find a significant interaction effect of the amyloid status and baseline cMD in predicting longitudinal tau in the entorhinal cortex (p = 0.044) but not the inferior temporal lobe, such that greater baseline cMD values predicts the accumulation of entorhinal tau in amyloid-positive participants. Moreover, we find a significant interaction effect of the amyloid status and baseline cMD in the entorhinal cortex (but not inferior temporal cMD) in predicting longitudinal LMDR (p < 0.001), such that baseline entorhinal cMD predicts the episodic memory decline in amyloid-positive participants. CONCLUSIONS The combination of amyloidosis and elevated cMD in the entorhinal cortex may help identify individuals at short-term risk of tau accumulation and Alzheimer's Disease-related episodic memory decline, suggesting utility in clinical trials.
Collapse
Affiliation(s)
- Geoffroy Gagliardi
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, 02129, USA
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Elena Rodriguez-Vieitez
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, 02129, USA
- Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, Stockholm, 14152, Sweden
| | - Victor Montal
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, 08041, Spain
- Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, 28031, Spain
| | - Jorge Sepulcre
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Gordon Center for Medical Imaging, Boston, MA, 02114, USA
| | - Ibai Diez
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Gordon Center for Medical Imaging, Boston, MA, 02114, USA
| | - Cristina Lois
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Gordon Center for Medical Imaging, Boston, MA, 02114, USA
| | - Bernard Hanseeuw
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Gordon Center for Medical Imaging, Boston, MA, 02114, USA
- Saint Luc University Hospital, Université Catholique de Louvain, Brussels, 1200, Belgium
| | - Aaron P Schultz
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, 02129, USA
| | - Michael J Properzi
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Kathryn V Papp
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, 02129, USA
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Gad A Marshall
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, 08041, Spain
- Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, 28031, Spain
| | - Keith A Johnson
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Gordon Center for Medical Imaging, Boston, MA, 02114, USA
| | - Reisa A Sperling
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, 02129, USA
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Patrizia Vannini
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, 02129, USA.
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
15
|
Pais MV, Forlenza OV, Diniz BS. Plasma Biomarkers of Alzheimer's Disease: A Review of Available Assays, Recent Developments, and Implications for Clinical Practice. J Alzheimers Dis Rep 2023; 7:355-380. [PMID: 37220625 PMCID: PMC10200198 DOI: 10.3233/adr-230029] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 04/03/2023] [Indexed: 05/25/2023] Open
Abstract
Recently, low-sensitive plasma assays have been replaced by new ultra-sensitive assays such as single molecule enzyme-linked immunosorbent assay (Simoa), the Mesoscale Discovery (MSD) platform, and immunoprecipitation-mass spectrometry (IP-MS) with higher accuracy in the determination of plasma biomarkers of Alzheimer's disease (AD). Despite the significant variability, many studies have established in-house cut-off values for the most promising available biomarkers. We first reviewed the most used laboratory methods and assays to measure plasma AD biomarkers. Next, we review studies focused on the diagnostic performance of these biomarkers to identify AD cases, predict cognitive decline in pre-clinical AD cases, and differentiate AD cases from other dementia. We summarized data from studies published until January 2023. A combination of plasma Aβ42/40 ratio, age, and APOE status showed the best accuracy in diagnosing brain amyloidosis with a liquid chromatography-mass spectrometry (LC-MS) assay. Plasma p-tau217 has shown the best accuracy in distinguishing Aβ-PET+ from Aβ-PET-even in cognitively unimpaired individuals. We also summarized the different cut-off values for each biomarker when available. Recently developed assays for plasma biomarkers have undeniable importance in AD research, with improved analytical and diagnostic performance. Some biomarkers have been extensively used in clinical trials and are now clinically available. Nonetheless, several challenges remain to their widespread use in clinical practice.
Collapse
Affiliation(s)
- Marcos V. Pais
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Faculdade de Medicina, Universidade de Sao Paulo (FMUSP), Sao Paulo, SP, Brazil
| | - Orestes V. Forlenza
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Faculdade de Medicina, Universidade de Sao Paulo (FMUSP), Sao Paulo, SP, Brazil
| | - Breno S. Diniz
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
16
|
Ali M, Archer DB, Gorijala P, Western D, Timsina J, Fernández MV, Wang TC, Satizabal CL, Yang Q, Beiser AS, Wang R, Chen G, Gordon B, Benzinger TLS, Xiong C, Morris JC, Bateman RJ, Karch CM, McDade E, Goate A, Seshadri S, Mayeux RP, Sperling RA, Buckley RF, Johnson KA, Won HH, Jung SH, Kim HR, Seo SW, Kim HJ, Mormino E, Laws SM, Fan KH, Kamboh MI, Vemuri P, Ramanan VK, Yang HS, Wenzel A, Rajula HSR, Mishra A, Dufouil C, Debette S, Lopez OL, DeKosky ST, Tao F, Nagle MW, Hohman TJ, Sung YJ, Dumitrescu L, Cruchaga C. Large multi-ethnic genetic analyses of amyloid imaging identify new genes for Alzheimer disease. Acta Neuropathol Commun 2023; 11:68. [PMID: 37101235 PMCID: PMC10134547 DOI: 10.1186/s40478-023-01563-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 04/05/2023] [Indexed: 04/28/2023] Open
Abstract
Amyloid PET imaging has been crucial for detecting the accumulation of amyloid beta (Aβ) deposits in the brain and to study Alzheimer's disease (AD). We performed a genome-wide association study on the largest collection of amyloid imaging data (N = 13,409) to date, across multiple ethnicities from multicenter cohorts to identify variants associated with brain amyloidosis and AD risk. We found a strong APOE signal on chr19q.13.32 (top SNP: APOE ɛ4; rs429358; β = 0.35, SE = 0.01, P = 6.2 × 10-311, MAF = 0.19), driven by APOE ɛ4, and five additional novel associations (APOE ε2/rs7412; rs73052335/rs5117, rs1081105, rs438811, and rs4420638) independent of APOE ɛ4. APOE ɛ4 and ε2 showed race specific effect with stronger association in Non-Hispanic Whites, with the lowest association in Asians. Besides the APOE, we also identified three other genome-wide loci: ABCA7 (rs12151021/chr19p.13.3; β = 0.07, SE = 0.01, P = 9.2 × 10-09, MAF = 0.32), CR1 (rs6656401/chr1q.32.2; β = 0.1, SE = 0.02, P = 2.4 × 10-10, MAF = 0.18) and FERMT2 locus (rs117834516/chr14q.22.1; β = 0.16, SE = 0.03, P = 1.1 × 10-09, MAF = 0.06) that all colocalized with AD risk. Sex-stratified analyses identified two novel female-specific signals on chr5p.14.1 (rs529007143, β = 0.79, SE = 0.14, P = 1.4 × 10-08, MAF = 0.006, sex-interaction P = 9.8 × 10-07) and chr11p.15.2 (rs192346166, β = 0.94, SE = 0.17, P = 3.7 × 10-08, MAF = 0.004, sex-interaction P = 1.3 × 10-03). We also demonstrated that the overall genetic architecture of brain amyloidosis overlaps with that of AD, Frontotemporal Dementia, stroke, and brain structure-related complex human traits. Overall, our results have important implications when estimating the individual risk to a population level, as race and sex will needed to be taken into account. This may affect participant selection for future clinical trials and therapies.
Collapse
Affiliation(s)
- Muhammad Ali
- Department of Psychiatry, Washington University, St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics, Washington University, St. Louis, MO, 63110, USA
| | - Derek B Archer
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Priyanka Gorijala
- Department of Psychiatry, Washington University, St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics, Washington University, St. Louis, MO, 63110, USA
| | - Daniel Western
- Department of Psychiatry, Washington University, St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics, Washington University, St. Louis, MO, 63110, USA
| | - Jigyasha Timsina
- Department of Psychiatry, Washington University, St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics, Washington University, St. Louis, MO, 63110, USA
| | - Maria V Fernández
- Department of Psychiatry, Washington University, St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics, Washington University, St. Louis, MO, 63110, USA
| | - Ting-Chen Wang
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Claudia L Satizabal
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health, San Antonio, TX, 78229, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Alexa S Beiser
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | | | - Gengsheng Chen
- Knight Alzheimer's Disease Research Center, Washington University, St Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University, St Louis, MO, USA
| | - Brian Gordon
- Knight Alzheimer's Disease Research Center, Washington University, St Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University, St Louis, MO, USA
| | - Tammie L S Benzinger
- Knight Alzheimer's Disease Research Center, Washington University, St Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University, St Louis, MO, USA
| | - Chengjie Xiong
- Knight Alzheimer's Disease Research Center, Washington University, St Louis, MO, USA
| | - John C Morris
- Knight Alzheimer's Disease Research Center, Washington University, St Louis, MO, USA
- Department of Neurology, Washington University, St Louis, MO, USA
| | - Randall J Bateman
- Knight Alzheimer's Disease Research Center, Washington University, St Louis, MO, USA
- Department of Neurology, Washington University, St Louis, MO, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Celeste M Karch
- Department of Psychiatry, Washington University, St. Louis, MO, 63110, USA
| | - Eric McDade
- Department of Neurology, Washington University, St Louis, MO, USA
| | - Alison Goate
- Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sudha Seshadri
- Framingham Heart Study, Framingham, MA, USA
- Boston University School of Medicine, Boston, MA, USA
| | - Richard P Mayeux
- The Department of Neurology, Columbia University, New York, NY, USA
| | - Reisa A Sperling
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Brigham and Women's Hospital and Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rachel F Buckley
- Brigham and Women's Hospital and Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Keith A Johnson
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hong-Hee Won
- Department of Digital Health, Samsung Medical Center, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Sang-Hyuk Jung
- Department of Digital Health, Samsung Medical Center, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Hang-Rai Kim
- Department of Neurology, Dongguk University Ilsan Hospital, Dongguk University College of Medicine, Goyang, Republic of Korea
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hee Jin Kim
- Department of Digital Health, Samsung Medical Center, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Elizabeth Mormino
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Simon M Laws
- Centre for Precision Health, Edith Cowan University, 270 Joondalup Dr, Joondalup, WA, 6027, Australia
| | - Kang-Hsien Fan
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - M Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Prashanthi Vemuri
- Department of Radiology, Mayo Clinic-Minnesota, Rochester, MN, 55905, USA
| | - Vijay K Ramanan
- Department of Neurology, Mayo Clinic-Minnesota, Rochester, MN, 55905, USA
| | - Hyun-Sik Yang
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, USA
| | - Allen Wenzel
- Wisconsin Alzheimer's Institute, Madison, WI, USA
| | - Hema Sekhar Reddy Rajula
- UMR 1219, University of Bordeaux, INSERM, Bordeaux Population Health Research Centre, Team ELEANOR, 33000, Bordeaux, France
| | - Aniket Mishra
- UMR 1219, University of Bordeaux, INSERM, Bordeaux Population Health Research Centre, Team ELEANOR, 33000, Bordeaux, France
| | - Carole Dufouil
- UMR 1219, University of Bordeaux, INSERM, Bordeaux Population Health Research Centre, Team ELEANOR, 33000, Bordeaux, France
| | - Stephanie Debette
- UMR 1219, University of Bordeaux, INSERM, Bordeaux Population Health Research Centre, Team ELEANOR, 33000, Bordeaux, France
- Department of Neurology, Boston University School of Medicine, Boston, MA, 2115, USA
- Department of Neurology, CHU de Bordeaux, 33000, Bordeaux, France
| | - Oscar L Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Steven T DeKosky
- Department of Neurology and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Feifei Tao
- Neurogenomics, Genetics-Guided Dementia Discovery, Eisai, Inc, Cambridge, MA, USA
| | - Michael W Nagle
- Neurogenomics, Genetics-Guided Dementia Discovery, Eisai, Inc, Cambridge, MA, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Yun Ju Sung
- Department of Psychiatry, Washington University, St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics, Washington University, St. Louis, MO, 63110, USA
| | - Logan Dumitrescu
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University, St. Louis, MO, 63110, USA.
- NeuroGenomics and Informatics, Washington University, St. Louis, MO, 63110, USA.
- Knight Alzheimer's Disease Research Center, Washington University, St Louis, MO, USA.
- Hope Center for Neurologic Diseases, Washington University, St. Louis, MO, 63110, USA.
- Department of Genetics, Washington University School of Medicine, St Louis, MO, 63110, USA.
| |
Collapse
|
17
|
Li JQ, Song JH, Suckling J, Wang YJ, Zuo CT, Zhang C, Gao J, Song YQ, Xie AM, Tan L, Yu JT. Disease trajectories in elders with suspected non-Alzheimer's pathophysiology and its comparison with Alzheimer's disease pathophysiology: a longitudinal study. RESEARCH SQUARE 2023:rs.3.rs-2744271. [PMID: 37034751 PMCID: PMC10081361 DOI: 10.21203/rs.3.rs-2744271/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
Background According to the new 'AT(N)' system, those with a normal amyloid biomarker but with abnormal tauopathy or biomarkers of neurodegeneration or neuronal injury, have been labeled suspected non-Alzheimer's pathophysiology (SNAP). We aimed to estimate the long-term clinical and cognitive trajectories of SNAP individuals in non-demented elders and its comparison with individual in the Alzheimer's disease (AD) pathophysiology using 'AT(N)' system. Methods We included individuals with available baseline cerebrospinal fluid (CSF) Aβ (A), CSF phosphorylated tau examination (T) and 18F-uorodeoxyglucose PET or volumetric magnetic resonance imaging (N) from the Alzheimer's Disease Neuroimaging Initiative database. Longitudinal change in clinical outcomes are assessed using linear mixed effects models. Conversion risk from cognitively normal (CN) to cognitively impairment, and conversion from mild cognitive impairment (MCI) to dementia are assessed using multivariate Cox proportional hazard models. Results Totally, 366 SNAP individuals were included (114 A-T-N-, 154 A-T + N-, 54 A-T-N + and 44 A-T + N+) of whom 178 were CN and 188 were MCI. Compared with A-T-N-, CN elders with A-T + N-, A-T-N + and A-T + N + had a faster rate of ADNI-MEM score decline. Moreover, CN older individuals with A-T + N + also had a faster rate of decline in ADNI-MEM score than those with A-T + N- individuals. MCI patients with A-T + N + had a faster rate of ADNI-MEM and ADNI-EF decline and hippocampal volume loss compared with A-T-N- and A-T + N- profiles. CN older individuals with A-T + N + had an increased risk of conversion to cognitive impairment (CDR-GS ≥ 0.5) compared with A-T + N- and A-T-N-. In MCI patients, A-T + N + also had an increased risk of conversion to dementia compared with A-T + N- and A-T-N-. Compared with A-T + N-, CN elders and MCI patients with A + T + N- and A + T + N + had a faster rate of ADNI-MEM score, ADNI-EF score decline, and hippocampal volume loss. CN individuals with A + T + N + had a faster rate of ADNI-EF score decline compare with A-T + N + individuals. Moreover, MCI patients with A + T + N + also had a faster rate of decline in ADNI-MEM score, ADNI-EF score and hippocampal volume loss than those with A-T + N + individuals. Conclusions The findings from clinical, imaging and biomarker studies on SNAP, and its comparison with AD pathophysiology offered an important foundation for future studies.
Collapse
Affiliation(s)
| | | | | | | | | | - Can Zhang
- Massachusetts General Hospital, Harvard Medical School
| | | | | | | | | | | |
Collapse
|
18
|
Jiao B, Li R, Zhou H, Qing K, Liu H, Pan H, Lei Y, Fu W, Wang X, Xiao X, Liu X, Yang Q, Liao X, Zhou Y, Fang L, Dong Y, Yang Y, Jiang H, Huang S, Shen L. Neural biomarker diagnosis and prediction to mild cognitive impairment and Alzheimer's disease using EEG technology. Alzheimers Res Ther 2023; 15:32. [PMID: 36765411 PMCID: PMC9912534 DOI: 10.1186/s13195-023-01181-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 02/06/2023] [Indexed: 02/12/2023]
Abstract
BACKGROUND Electroencephalogram (EEG) has emerged as a non-invasive tool to detect the aberrant neuronal activity related to different stages of Alzheimer's disease (AD). However, the effectiveness of EEG in the precise diagnosis and assessment of AD and its preclinical stage, amnestic mild cognitive impairment (MCI), has yet to be fully elucidated. In this study, we aimed to identify key EEG biomarkers that are effective in distinguishing patients at the early stage of AD and monitoring the progression of AD. METHODS A total of 890 participants, including 189 patients with MCI, 330 patients with AD, 125 patients with other dementias (frontotemporal dementia, dementia with Lewy bodies, and vascular cognitive impairment), and 246 healthy controls (HC) were enrolled. Biomarkers were extracted from resting-state EEG recordings for a three-level classification of HC, MCI, and AD. The optimal EEG biomarkers were then identified based on the classification performance. Random forest regression was used to train a series of models by combining participants' EEG biomarkers, demographic information (i.e., sex, age), CSF biomarkers, and APOE phenotype for assessing the disease progression and individual's cognitive function. RESULTS The identified EEG biomarkers achieved over 70% accuracy in the three-level classification of HC, MCI, and AD. Among all six groups, the most prominent effects of AD-linked neurodegeneration on EEG metrics were localized at parieto-occipital regions. In the cross-validation predictive analyses, the optimal EEG features were more effective than the CSF + APOE biomarkers in predicting the age of onset and disease course, whereas the combination of EEG + CSF + APOE measures achieved the best performance for all targets of prediction. CONCLUSIONS Our study indicates that EEG can be used as a useful screening tool for the diagnosis and disease progression evaluation of MCI and AD.
Collapse
Affiliation(s)
- Bin Jiao
- grid.216417.70000 0001 0379 7164Department of Neurology, Xiangya Hospital, Central South University, Changsha, China ,grid.216417.70000 0001 0379 7164National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China ,grid.216417.70000 0001 0379 7164Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China ,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China ,grid.216417.70000 0001 0379 7164Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Rihui Li
- grid.168010.e0000000419368956Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA USA ,Brainup Institute of Science and Technology, Chongqing, China
| | - Hui Zhou
- grid.216417.70000 0001 0379 7164Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Kunqiang Qing
- Brainup Institute of Science and Technology, Chongqing, China
| | - Hui Liu
- grid.216417.70000 0001 0379 7164Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Hefu Pan
- Brainup Institute of Science and Technology, Chongqing, China
| | - Yanqin Lei
- Brainup Institute of Science and Technology, Chongqing, China
| | - Wenjin Fu
- Brainup Institute of Science and Technology, Chongqing, China
| | - Xiaoan Wang
- Brainup Institute of Science and Technology, Chongqing, China
| | - Xuewen Xiao
- grid.216417.70000 0001 0379 7164Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xixi Liu
- grid.216417.70000 0001 0379 7164Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Qijie Yang
- grid.216417.70000 0001 0379 7164Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xinxin Liao
- grid.216417.70000 0001 0379 7164Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Yafang Zhou
- grid.216417.70000 0001 0379 7164Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Liangjuan Fang
- grid.216417.70000 0001 0379 7164Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yanbin Dong
- Brainup Institute of Science and Technology, Chongqing, China
| | - Yuanhao Yang
- grid.1003.20000 0000 9320 7537Mater Research Institute, The University of Queensland, Woolloongabba, Queensland 4102 Australia
| | - Haiyan Jiang
- grid.216417.70000 0001 0379 7164Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Sha Huang
- grid.216417.70000 0001 0379 7164Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China. .,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China. .,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China. .,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China. .,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China. .,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.
| |
Collapse
|
19
|
Imbimbo BP, Ippati S, Watling M, Imbimbo C. Role of monomeric amyloid-β in cognitive performance in Alzheimer's disease: Insights from clinical trials with secretase inhibitors and monoclonal antibodies. Pharmacol Res 2023; 187:106631. [PMID: 36586644 DOI: 10.1016/j.phrs.2022.106631] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/18/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
According to the β-amyloid (Aβ) hypothesis of Alzheimer's disease (AD), brain Aβ accumulation is the primary cascade event leading to cognitive deficit and dementia. Numerous anti-Aβ drugs either inhibiting production or aggregation of Aβ or stimulating its clearance have failed to show clinical benefit in large scale AD trials, with β- and γ-secretase inhibitors consistently worsening cognitive and clinical decline. In June 2021, the FDA approved aducanumab, an anti-Aβ monoclonal antibody for early AD based on its ability to reduce brain amyloid plaques, while two other amyloid-clearing antibodies (lecanemab and donanemab) have recently produced encouraging cognitive and clinical results. We reviewed AD trials using PubMed, meeting abstracts and ClinicalTrials.gov and evaluated the effects of such drugs on cerebrospinal fluid (CSF) Aβ levels, correlating them with cognitive effects. We found that β-secretase and γ-secretase inhibitors produce detrimental cognitive effects by significantly reducing CSF Aβ levels. We speculate that monoclonal antibodies targeting Aβ protofibrils, fibrils or plaques may improve cognitive performance in early AD by increasing soluble Aβ levels through Aβ aggregate disassembly and/or stabilization of existing Aβ monomers.These findings suggest that the real culprit in AD may be decreased levels of soluble monomeric Aβ due to sequestration into brain Aβ aggregates and plaques.
Collapse
Affiliation(s)
- Bruno P Imbimbo
- Department of Research & Development, Chiesi Farmaceutici, Parma, Italy.
| | - Stefania Ippati
- San Raffaele Scientific Institute, San Raffaele Hospital, 20132 Milan, Italy
| | - Mark Watling
- CNS & Pain Department, TranScrip Ltd, Reading, UK
| | - Camillo Imbimbo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
20
|
Haas AL, Olm P, Utz J, Siegmann EM, Spitzer P, Florvaag A, Schmidt MA, Doerfler A, Lewczuk P, Kornhuber J, Maler JM, Oberstein TJ. PASSED: Brain atrophy in non-demented individuals in a long-term longitudinal study from two independent cohorts. Front Aging Neurosci 2023; 15:1121500. [PMID: 36909947 PMCID: PMC9992803 DOI: 10.3389/fnagi.2023.1121500] [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: 12/11/2022] [Accepted: 01/31/2023] [Indexed: 02/25/2023] Open
Abstract
Introduction Alzheimer's disease (AD) is indicated by a decrease in amyloid beta 42 (Aβ42) level or the Aβ42/Aβ40 ratio, and by increased levels of Tau with phosphorylated threonine at position 181 (pTau181) in cerebrospinal fluid (CSF) years before the onset of clinical symptoms. However, once only pTau181 is increased, cognitive decline in individuals with subjective or mild cognitive impairment is slowed compared to individuals with AD. Instead of a decrease in Aβ42 levels, an increase in Aβ42 was observed in these individuals, leading to the proposal to refer to them as nondemented subjects with increased pTau-levels and Aβ surge with subtle cognitive deterioration (PASSED). In this study, we determined the longitudinal atrophy rates of AD, PASSED, and Biomarker-negative nondemented individuals of two independent cohorts to determine whether these groups can be distinguished by their longitudinal atrophy patterns or rates. Methods Depending on their CSF-levels of pTau 181 (T), total Tau (tTau, N), Aβ42 or ratio of Aβ42/Aβ40 (A), 185 non-demented subjects from the Alzheimer's Disease Neuroimaging Initiative (ADNI) and 62 non-demented subjects from Erlangen AD cohort were assigned to an ATN group (A-T-N-, A-T+N±, A+T-N±and A+T+N±) and underwent T1-weighted structural magnetic resonance imaging (sMRI). Longitudinal grey matter (GM) atrophy patterns were assessed with voxel-based morphometry (VBM) using the cat12 toolbox on spm12 (statistical parametric mapping) of MRI scans from individuals in the ADNI cohort with a mean follow-up of 2 and 5 years, respectively. The annualized atrophy rate for individuals in the Erlangen cohort was determined using region of interest analysis (ROI) in terms of a confirmatory analysis. Results In the A-T+N± group, VBM did not identify any brain region that showed greater longitudinal atrophy than the A+T+N±, A+T+N± or biomarker negative control group. In contrast, marked longitudinal atrophy in the temporal lobe was evident in the A+T-N± group compared with A+T-N± and biomarker-negative subjects. The ROI in the angular gyrus identified by VBM analysis of the ADNI cohort did not discriminate better than the hippocampal volume and atrophy rate between AD and PASSED in the confirmatory analysis. Discussion In this study, nondemented subjects with PASSED did not show a unique longitudinal atrophy pattern in comparison to nondemented subjects with AD. The nonsignificant atrophy rate compared with controls suggests that increased pTau181-levels without concomitant amyloidopathy did not indicate a neurodegenerative disorder.
Collapse
Affiliation(s)
- Anna-Lena Haas
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Pauline Olm
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Janine Utz
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Eva-Maria Siegmann
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Philipp Spitzer
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Anna Florvaag
- Institute of Neuroradiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Manuel Alexander Schmidt
- Institute of Neuroradiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Arnd Doerfler
- Institute of Neuroradiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Neurodegeneration Diagnostics, Department of Biochemical Diagnostics, University Hospital of Bialystok, Medical University of Bialystok, Bialystok, Poland
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Juan Manuel Maler
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Timo Jan Oberstein
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | |
Collapse
|
21
|
Utz J, Olm P, Jablonowski J, Siegmann EM, Spitzer P, Lewczuk P, Kornhuber J, Maler JM, Oberstein TJ. Reconceptualization of the Erlangen Score for the Assessment of Dementia Risk: The ERlangen Score. J Alzheimers Dis 2023; 96:265-275. [PMID: 37742651 PMCID: PMC10657695 DOI: 10.3233/jad-230524] [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] [Accepted: 08/17/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND The established Erlangen Score (ES) for the interpretation of cerebrospinal fluid (CSF) biomarkers in the diagnostics of Alzheimer's disease (AD) uses markers of amyloidopathy and tauopathy, equally weighted to form an easy-interpretable ordinal scale. However, these biomarkers are not equally predictive for AD. OBJECTIVE The higher weighting of the Aβ42/Aβ40 ratio, as a reconceptualized ERlangen Score (ERS), was tested for advantages in diagnostic performance. METHODS Non-demented subjects (N = 154) with a mean follow up of 5 years were assigned to a group ranging from 0 to 4 in ES or ERS. Psychometric trajectories and dementia risk were assessed. RESULTS The distribution of subjects between ES and ERS among the groups differed considerably, as grouping allocated 32 subjects to ES group 2, but only 2 to ERS group 2. The discriminative accuracy between the ES (AUC 73.2%, 95% CI [64.2, 82.2]) and ERS (AUC 72.0%, 95% CI [63.1, 81.0]) for dementia risk showed no significant difference. Without consideration of the Aβ42/Aβ40 ratio in ES grouping, the optimal cut-off of the ES shifted to ≥2. CONCLUSIONS The ERS showed advantages over the ES in test interpretation with comparable overall test performance, as fewer cases were allocated to the intermediate risk group. The established cut-off of ≥2 can be maintained for the ERS, whereas it must be adjusted for the ES when determining the Aβ42/Aβ40 ratio.
Collapse
Affiliation(s)
- Janine Utz
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Pauline Olm
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Johannes Jablonowski
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Eva-Maria Siegmann
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Philipp Spitzer
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, and Department of Biochemical Diagnostics, University Hospital of Bialystok, Białystok, Poland
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Juan Manuel Maler
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Timo Jan Oberstein
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| |
Collapse
|
22
|
Fowler CJ, Stoops E, Rainey‐Smith SR, Vanmechelen E, Vanbrabant J, Dewit N, Mauroo K, Maruff P, Rowe CC, Fripp J, Li Q, Bourgeat P, Collins SJ, Martins RN, Masters CL, Doecke JD. Plasma p-tau181/Aβ 1-42 ratio predicts Aβ-PET status and correlates with CSF-p-tau181/Aβ 1-42 and future cognitive decline. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12375. [PMID: 36447478 PMCID: PMC9695763 DOI: 10.1002/dad2.12375] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 09/28/2022] [Indexed: 11/26/2022]
Abstract
Background In Alzheimer's disease (AD), plasma amyloid beta (Aβ)1-42 and phosphorylated tau (p-tau) predict high amyloid status from Aβ positron emission tomography (PET); however, the extent to which combination of these plasma assays can predict remains unknown. Methods Prototype Simoa assays were used to measure plasma samples from participants who were either cognitively normal (CN) or had mild cognitive impairment (MCI)/AD in the Australian Imaging, Biomarkers and Lifestyle (AIBL) study. Results The p-tau181/Aβ1-42 ratio showed the best prediction of Aβ-PET across all participants (area under the curve [AUC] = 0.905, 95% confidence interval [CI]: 0.86-0.95) and in CN (AUC = 0.873; 0.80-0.94), and symptomatic (AUC = 0.908; 0.82-1.00) adults. Plasma p-tau181/Aβ1-42 ratio correlated with cerebrospinal fluid (CSF) p-tau181 (Elecsys, Spearman's ρ = 0.74, P < 0.0001) and predicted abnormal CSF Aβ (AUC = 0.816; 0.74-0.89). The p-tau181/Aβ1-42 ratio also predicted future rates of cognitive decline assessed by AIBL Preclinical Alzheimer Cognitive Composite or Clinical Dementia Rating Sum of Boxes (P < 0.0001). Discussion Plasma p-tau181/Aβ1-42 ratio predicted both Aβ-PET status and cognitive decline, demonstrating potential as both a diagnostic aid and as a screening and prognostic assay for preclinical AD trials.
Collapse
Affiliation(s)
| | | | - Stephanie R. Rainey‐Smith
- School of Medical and Health SciencesCentre of Excellence for Alzheimer's Disease Research & CareEdith Cowan UniversityJoondalupWestern AustraliaAustralia
| | | | | | | | | | | | - Christopher C. Rowe
- The Florey Institute of Neuroscience and Mental HealthMelbourneVictoriaAustralia
- Austin Health, Molecular Imaging Researchand The Florey Department of NeuroscienceUniversity of MelbourneMelbourneVictoriaAustralia
| | - Jurgen Fripp
- Australian E‐Health Research CentreCSIROHerstonQueenslandAustralia
| | - Qiao‐Xin Li
- The Florey Institute of Neuroscience and Mental HealthMelbourneVictoriaAustralia
| | | | - Steven J. Collins
- Department of Medicine (RMH)The University of MelbourneMelbourneVictoriaAustralia
| | - Ralph N. Martins
- School of Medical and Health SciencesCentre of Excellence for Alzheimer's Disease Research & CareEdith Cowan UniversityJoondalupWestern AustraliaAustralia
- Department of Biological SciencesMacquarie UniversityNorth RydeNew South WalesAustralia
| | - Colin L. Masters
- The Florey Institute of Neuroscience and Mental HealthMelbourneVictoriaAustralia
| | - James D. Doecke
- Australian E‐Health Research CentreCSIROHerstonQueenslandAustralia
| |
Collapse
|
23
|
Chang HI, Hsu SW, Kao ZK, Lee CC, Huang SH, Lin CH, Liu MN, Chang CC. Impact of Amyloid Pathology in Mild Cognitive Impairment Subjects: The Longitudinal Cognition and Surface Morphometry Data. Int J Mol Sci 2022; 23:ijms232314635. [PMID: 36498962 PMCID: PMC9738566 DOI: 10.3390/ijms232314635] [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: 09/05/2022] [Revised: 11/13/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022] Open
Abstract
The amyloid framework forms the central medical theory related to Alzheimer disease (AD), and the in vivo demonstration of amyloid positivity is essential for diagnosing AD. On the basis of a longitudinal cohort design, the study investigated clinical progressive patterns by obtaining cognitive and structural measurements from a group of patients with amnestic mild cognitive impairment (MCI); the measurements were classified by the positivity (Aβ+) or absence (Aβ-) of the amyloid biomarker. We enrolled 185 patients (64 controls, 121 patients with MCI). The patients with MCI were classified into two groups on the basis of their [18F]flubetaben or [18F]florbetapir amyloid positron-emission tomography scan (Aβ+ vs. Aβ-, 67 vs. 54 patients) results. Data from annual cognitive measurements and three-dimensional T1 magnetic resonance imaging scans were used for between-group comparisons. To obtain longitudinal cognitive test scores, generalized estimating equations were applied. A linear mixed effects model was used to compare the time effect of cortical thickness degeneration. The cognitive decline trajectory of the Aβ+ group was obvious, whereas the Aβ- and control groups did not exhibit a noticeable decline over time. The group effects of cortical thickness indicated decreased entorhinal cortex in the Aβ+ group and supramarginal gyrus in the Aβ- group. The topology of neurodegeneration in the Aβ- group was emphasized in posterior cortical regions. A comparison of the changes in the Aβ+ and Aβ- groups over time revealed a higher rate of cortical thickness decline in the Aβ+ group than in the Aβ- group in the default mode network. The Aβ+ and Aβ- groups experienced different APOE ε4 effects. For cortical-cognitive correlations, the regions associated with cognitive decline in the Aβ+ group were mainly localized in the perisylvian and anterior cingulate regions. By contrast, the degenerative topography of Aβ- MCI was scattered. The memory learning curves, cognitive decline patterns, and cortical degeneration topographies of the two MCI groups were revealed to be different, suggesting a difference in pathophysiology. Longitudinal analysis may help to differentiate between these two MCI groups if biomarker access is unavailable in clinical settings.
Collapse
Affiliation(s)
- Hsin-I Chang
- Department of Neurology, Cognition and Aging Center, Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Shih-Wei Hsu
- Department of Radiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Zih-Kai Kao
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chen-Chang Lee
- Department of Radiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Shu-Hua Huang
- Department of Nuclear Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Ching-Heng Lin
- Center for Artificial Intelligence in Medicine, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- Bachelor Program in Artificial Intelligence, Chang Gung University, Taoyuan 333, Taiwan
| | - Mu-N Liu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei 112, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Correspondence: (M.-N.L.); (C.-C.C.)
| | - Chiung-Chih Chang
- Department of Neurology, Cognition and Aging Center, Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Correspondence: (M.-N.L.); (C.-C.C.)
| |
Collapse
|
24
|
Vipin A, Koh CL, Wong BYX, Zailan FZ, Tan JY, Soo SA, Satish V, Kumar D, Wang BZ, Ng ASL, Chiew HJ, Ng KP, Kandiah N. Amyloid-Tau-Neurodegeneration Profiles and Longitudinal Cognition in Sporadic Young-Onset Dementia. J Alzheimers Dis 2022; 90:543-551. [DOI: 10.3233/jad-220448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We examined amyloid-tau-neurodegeneration biomarker effects on cognition in a Southeast-Asian cohort of 84 sporadic young-onset dementia (YOD; age-at-onset <65 years) patients. They were stratified into A+N+, A– N+, and A– N– profiles via cerebrospinal fluid amyloid-β1–42 (A), phosphorylated-tau (T), MRI medial temporal atrophy (neurodegeneration– N), and confluent white matter hyperintensities cerebrovascular disease (CVD). A, T, and CVD effects on longitudinal Mini-Mental State Examination (MMSE) were evaluated. A+N+ patients demonstrated steeper MMSE decline than A– N+ (β = 1.53; p = 0.036; CI 0.15:2.92) and A– N– (β = 4.68; p = 0.001; CI 1.98:7.38) over a mean follow-up of 1.24 years. Within A– N+, T– CVD+ patients showed greater MMSE decline compared to T+CVD– patients (β = – 2.37; p = 0.030; CI – 4.41:– 0.39). A+ results in significant cognitive decline, while CVD influences longitudinal cognition in the A– sub-group.
Collapse
Affiliation(s)
- Ashwati Vipin
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
- National Neuroscience Institute, Singapore, Singapore
| | - Chen Ling Koh
- National Neuroscience Institute, Singapore, Singapore
| | | | - Fatin Zahra Zailan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
- National Neuroscience Institute, Singapore, Singapore
| | - Jayne Yi Tan
- National Neuroscience Institute, Singapore, Singapore
| | - See Ann Soo
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
- National Neuroscience Institute, Singapore, Singapore
| | - Vaynii Satish
- National Neuroscience Institute, Singapore, Singapore
| | - Dilip Kumar
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
- National Neuroscience Institute, Singapore, Singapore
| | | | - Adeline Su Lyn Ng
- National Neuroscience Institute, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Hui Jin Chiew
- National Neuroscience Institute, Singapore, Singapore
| | - Kok Pin Ng
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
- National Neuroscience Institute, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Nagaendran Kandiah
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
- National Neuroscience Institute, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
25
|
Baldeiras I, Silva-Spínola A, Lima M, Leitão MJ, Durães J, Vieira D, Tbuas-Pereira M, Cruz VT, Rocha R, Alves L, Machado Á, Milheiro M, Santiago B, Santana I. Alzheimer’s Disease Diagnosis Based on the Amyloid, Tau, and Neurodegeneration Scheme (ATN) in a Real-Life Multicenter Cohort of General Neurological Centers. J Alzheimers Dis 2022; 90:419-432. [DOI: 10.3233/jad-220587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: The ATN scheme was proposed as an unbiased biological characterization of the Alzheimer’s disease (AD) spectrum, grouping biomarkers into three categories: brain Amyloidosis-A, Tauopathy-T, Neurodegeneration-N. Although this scheme was mainly recommended for research, it is relevant for diagnosis. Objective: To evaluate the ATN scheme performance in real-life cohorts reflecting the inflow of patients with cognitive complaints and different underlying disorders in general neurological centers. Methods: We included patients (n = 1,128) from six centers with their core cerebrospinal fluid-AD biomarkers analyzed centrally. A was assessed through Aβ 42/Aβ 40, T through pTau-181, and N through tTau. Association between demographic features, clinical diagnosis at baseline/follow-up and ATN profiles was assessed. Results: The prevalence of ATN categories was: A-T-N-: 28.3% ; AD continuum (A + T-/+N-/+): 47.8% ; non-AD (A- plus T or/and N+): 23.9% . ATN profiles prevalence was strongly influenced by age, showing differences according to gender, APOE genotype, and cognitive status. At baseline, 74.6% of patients classified as AD fell in the AD continuum, decreasing to 47.4% in mild cognitive impairment and 42.3% in other neurodegenerative conditions. At follow-up, 41% of patients changed diagnosis, and 92% of patients that changed to AD were classified within the AD continuum. A + was the best individual marker for predicting a final AD diagnosis, and the combinations A + T+(irrespective of N) and A + T+N+had the highest overall accuracy (83%). Conclusion: The ATN scheme is useful to guide AD diagnosis real-life neurological centers settings. However, it shows a lack of accuracy for patients with other types of dementia. In such cases, the inclusion of other markers specific for non-AD proteinopathies could be an important aid to the differential diagnosis.
Collapse
Affiliation(s)
- Inês Baldeiras
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Anuschka Silva-Spínola
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Marisa Lima
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Center for Research in Neuropsychology and Cognitive Behavioral Intervention, Faculty of Psychology and Educational Sciences, University of Coimbra, Coimbra, Portugal
| | - Maria João Leitão
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - João Durães
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Daniela Vieira
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Miguel Tbuas-Pereira
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | | | - Raquel Rocha
- ULSM Unidade Local de Sáude de Matosinhos, Matosinhos, Portugal
| | - Luisa Alves
- Hospital Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Lisboa, Portugal
| | | | | | | | - Isabel Santana
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
26
|
Ebenau JL, Visser D, Kroeze LA, van Leeuwenstijn MSSA, van Harten AC, Windhorst AD, Golla SVS, Boellaard R, Scheltens P, Barkhof F, van Berckel BNM, van der Flier WM. Longitudinal change in ATN biomarkers in cognitively normal individuals. Alzheimers Res Ther 2022; 14:124. [PMID: 36057616 PMCID: PMC9440493 DOI: 10.1186/s13195-022-01069-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/23/2022] [Indexed: 04/14/2023]
Abstract
BACKGROUND Biomarkers for amyloid, tau, and neurodegeneration (ATN) have predictive value for clinical progression, but it is not clear how individuals move through these stages. We examined changes in ATN profiles over time, and investigated determinants of change in A status, in a sample of cognitively normal individuals presenting with subjective cognitive decline (SCD). METHODS We included 92 individuals with SCD from the SCIENCe project with [18F]florbetapir PET (A) available at two time points (65 ± 8y, 42% female, MMSE 29 ± 1, follow-up 2.5 ± 0.7y). We additionally used [18F]flortaucipir PET for T and medial temporal atrophy score on MRI for N. Thirty-nine individuals had complete biomarker data at baseline and follow-up, enabling the construction of ATN profiles at two time points. All underwent extensive neuropsychological assessments (follow-up time 4.9 ± 2.8y, median number of visits n = 4). We investigated changes in biomarker status and ATN profiles over time. We assessed which factors predisposed for a change from A- to A+ using logistic regression. We additionally used linear mixed models to assess change from A- to A+, compared to the group that remained A- at follow-up, as predictor for cognitive decline. RESULTS At baseline, 62% had normal AD biomarkers (A-T-N- n = 24), 5% had non-AD pathologic change (A-T-N+ n = 2,) and 33% fell within the Alzheimer's continuum (A+T-N- n = 9, A+T+N- n = 3, A+T+N+ n = 1). Seventeen subjects (44%) changed to another ATN profile over time. Only 6/17 followed the Alzheimer's disease sequence of A → T → N, while 11/17 followed a different order (e.g., reverted back to negative biomarker status). APOE ε4 carriership inferred an increased risk of changing from A- to A+ (OR 5.2 (95% CI 1.2-22.8)). Individuals who changed from A- to A+, showed subtly steeper decline on Stroop I (β - 0.03 (SE 0.01)) and Stroop III (- 0.03 (0.01)), compared to individuals who remained A-. CONCLUSION We observed considerable variability in the order of ATN biomarkers becoming abnormal. Individuals who became A+ at follow-up showed subtle decline on tests for attention and executive functioning, confirming clinical relevance of amyloid positivity.
Collapse
Affiliation(s)
- Jarith L Ebenau
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands.
| | - Denise Visser
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Lior A Kroeze
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Mardou S S A van Leeuwenstijn
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 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
| | - Albert D Windhorst
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Sandeep V S Golla
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Frederik Barkhof
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- UCL Institutes of Neurology and Healthcare Engineering, London, UK
| | - Bart N M van Berckel
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Epidemiology & Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| |
Collapse
|
27
|
Oberstein TJ, Schmidt MA, Florvaag A, Haas AL, Siegmann EM, Olm P, Utz J, Spitzer P, Doerfler A, Lewczuk P, Kornhuber J, Maler JM. Amyloid-β levels and cognitive trajectories in non-demented pTau181-positive subjects without amyloidopathy. Brain 2022; 145:4032-4041. [PMID: 35973034 DOI: 10.1093/brain/awac297] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/11/2022] [Accepted: 07/24/2022] [Indexed: 11/13/2022] Open
Abstract
Phosphorylated Tau181 (pTau181) in cerebrospinal fluid (CSF) and recently in plasma has been associated with Alzheimer's disease. In the absence of amyloidopathy, individuals with increased total Tau levels and/or temporal lobe atrophy experience no or only mild cognitive decline compared with biomarker-negative controls, leading to the proposal to categorize this constellation as Suspected non-Alzheimer disease pathophysiology (SNAP). We investigated whether the characteristics of SNAP also applied to individuals with increased CSF-pTau181 without amyloidopathy. In this long-term observational study, 285 non-demented individuals, including 76 individuals with subjective cognitive impairment and 209 individuals with mild cognitive impairment, were classified based on their CSF-levels of pTau181 (T), total Tau (N), Amyloid-beta-(Aβ)-42 and Aβ42/Aβ40 ratio (A) into A + T+N±, A + T-N±, A-T + N±, and A-T-N-. The longitudinal analysis included 154 subjects with a follow-up of more than 12 months who were followed to a median of 4.6 years (interquartile range = 4.3 years). We employed linear mixed models on psychometric tests and region of interest analysis of structural MRI data. Cognitive decline and hippocampal atrophy rate were significantly higher in A + T+N ± compared to A-T + N±, whereas there was no difference between A-T + N ± and A-T-N-. Furthermore, there was no significant difference between A-T + N ± and controls in dementia risk (Hazard ratio 0.3, 95% confidence interval [0.1, 1.9]). However, A-T + N ± and A-T-N- could be distinguished based on their Aβ42 and Aβ40 levels. Both Aβ40 and Aβ42 levels were significantly increased in A-T + N ± compared to controls. Long term follow-up of A-T + N ± individuals revealed no evidence that this biomarker constellation was associated with dementia or more severe hippocampal atrophy rates compared to controls. However, because of the positive association of pTau181 with Aβ in the A-T + N ± group, a link to the pathophysiology of Alzheimer´s disease cannot be excluded in this case. We propose to refer to these individuals in the SNAP group as "pTau and Aβ surge with subtle deterioration" (PASSED). The investigation of the circumstances of simultaneous elevation of pTau and Aβ might provide a deeper insight into the process under which Aβ becomes pathological.
Collapse
Affiliation(s)
- Timo Jan Oberstein
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Manuel Alexander Schmidt
- Institute of Neuroradiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Anna Florvaag
- Department of Radiology and Nuclear Medicine, Klinikum Nuremberg, Nuremberg, Germany
| | - Anna Lena Haas
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Eva Maria Siegmann
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Pauline Olm
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Janine Utz
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Philipp Spitzer
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Arnd Doerfler
- Institute of Neuroradiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Neurodegeneration Diagnostics, Medical University of Bialystok, University Hospital of Bialystok, Bialystok, Poland.,Department of Biochemical Diagnostics, University Hospital of Bialystok, Bialystok, Poland
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Juan Manuel Maler
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
28
|
Tondelli M, Salemme S, Vinceti G, Bedin R, Trenti T, Molinari MA, Chiari A, Zamboni G. Predictive value of phospho-tau/total-tau ratio in amyloid-negative Mild Cognitive Impairment. Neurosci Lett 2022; 787:136811. [PMID: 35870715 DOI: 10.1016/j.neulet.2022.136811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/12/2022] [Accepted: 07/17/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND In patients with Mild Cognitive Impairment and normal biomarkers of amyloid-β deposition, prognostication remains challenging. METHODS We aimed at identifying clinical features, patterns of brain atrophy, and risk of subsequent conversion to dementia in a clinical cohort of consecutive patients with Mild Cognitive Impairment and normal CSF amyloid-β1-42 presenting to our Cognitive Neurology Clinic who were followed prospectively over an average of 25 months. We stratified them as Converters/Non-Converters to dementia based on clinical follow-up and compared baseline clinical features, CSF biomarkers, and pattern of atrophy on MRI data between groups. RESULTS Among 111 eligible patients (mean age 65,61 years; 56,8% were male), 41 patients developed a clinical diagnosis of dementia. Subjects with low baseline p/t-tau had twofold risk of future conversion compared to high p/t-tau ratio subjects (HR = 2.0, p = 0.026). When stratifying converters according to CSF p/t-tau ratio cut off value (0,17), those with values lower than the cut-off had significantly more MRI atrophy at baseline relative to Non-Converters in limbic structures. CONCLUSION In Mild Cognitive Impairment patients with negative CSF amyloid biomarker, CSF p/t-tau ratio may be useful to identify those at greater risk of subsequent conversion, possibly because of TDP43-related underlying pathology.
Collapse
Affiliation(s)
- Manuela Tondelli
- Department of Biomedical, Metabolic, and Neural Science, University of Modena and Reggio Emilia, Modena, Italy; Center for Neurosciences and Neurotechnology, Università di Modena e Reggio Emilia, Italy
| | - Simone Salemme
- Department of Biomedical, Metabolic, and Neural Science, University of Modena and Reggio Emilia, Modena, Italy; Center for Neurosciences and Neurotechnology, Università di Modena e Reggio Emilia, Italy
| | - Giulia Vinceti
- Department of Biomedical, Metabolic, and Neural Science, University of Modena and Reggio Emilia, Modena, Italy; Center for Neurosciences and Neurotechnology, Università di Modena e Reggio Emilia, Italy; Neurology Unit, Baggiovara Hospital, AOU Modena, Italy
| | - Roberta Bedin
- Department of Biomedical, Metabolic, and Neural Science, University of Modena and Reggio Emilia, Modena, Italy; Center for Neurosciences and Neurotechnology, Università di Modena e Reggio Emilia, Italy
| | - Tommaso Trenti
- Laboratory Medicine Department, Baggiovara Hospital, AOU Modena, Italy
| | | | | | - Giovanna Zamboni
- Department of Biomedical, Metabolic, and Neural Science, University of Modena and Reggio Emilia, Modena, Italy; Center for Neurosciences and Neurotechnology, Università di Modena e Reggio Emilia, Italy; Neurology Unit, Baggiovara Hospital, AOU Modena, Italy; Nuffield Department of Clinical Neurosciences, University of Oxford, UK.
| |
Collapse
|
29
|
Krishnadas N, Huang K, Schultz SA, Doré V, Bourgeat P, Goh AM, Lamb F, Bozinovski S, Burnham SC, Robertson JS, Laws SM, Maruff P, Masters CL, Villemagne VL, Rowe CC. Visually Identified Tau 18F-MK6240 PET Patterns in Symptomatic Alzheimer’s Disease. J Alzheimers Dis 2022; 88:1627-1637. [PMID: 35811517 PMCID: PMC9484111 DOI: 10.3233/jad-215558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: In Alzheimer’s disease, heterogeneity has been observed in the postmortem distribution of tau neurofibrillary tangles. Visualizing the topography of tau in vivo may facilitate clinical trials and clinical practice. Objective: This study aimed to investigate whether tau distribution patterns that are limited to mesial temporal lobe (MTL)/limbic regions, and those that spare MTL regions, can be visually identified using 18F-MK6240, and whether these patterns are associated with different demographic and cognitive profiles. Methods: Tau 18F-MK6240 PET images of 151 amyloid-β positive participants with mild cognitive impairment (MCI) and dementia were visually rated as: tau negative, limbic predominant (LP), MTL-sparing, and Typical by two readers. Groups were evaluated for differences in age, APOE ɛ4 carriage, hippocampal volumes, and cognition (MMSE, composite memory and non-memory scores). Voxel-wise contrasts were also performed. Results: Visual rating resulted in 59.6% classified as Typical, 17.9% as MTL-sparing, 9.9% LP, and 12.6% as tau negative. Intra-rater and inter-rater reliability was strong (Cohen’s kappa values of 0.89 and 0.86 respectively). Tracer retention in a “hook”-like distribution on sagittal sequences was observed in the LP and Typical groups. The visually classified MTL-sparing group had lower APOE ɛ4 carriage and relatively preserved hippocampal volumes. Higher MTL tau was associated with greater amnestic cognitive impairment. High cortical tau was associated with greater impairments on non-memory domains of cognition, and individuals with high cortical tau were more likely to have dementia than MCI. Conclusion: Tau distribution patterns can be visually identified using 18F-MK6240 PET and are associated with differences in APOE ɛ4 carriage, hippocampal volumes, and cognition.
Collapse
Affiliation(s)
- Natasha Krishnadas
- Florey Department of Neurosciences & Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Kun Huang
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Stephanie A. Schultz
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Vincent Doré
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC, Australia
- Health and Biosecurity Flagship, The Australian eHealth Research Centre, Melbourne, Victoria, Australia
| | - Pierrick Bourgeat
- Health and Biosecurity Flagship, The Australian eHealth Research Centre, Brisbane, QLD, Australia
| | - Anita M.Y. Goh
- Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia
- National Ageing Research Institute, Parkville, VIC, Australia
| | - Fiona Lamb
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Svetlana Bozinovski
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Samantha C. Burnham
- Health and Biosecurity Flagship, The Australian eHealth Research Centre, Melbourne, Victoria, Australia
| | - Joanne S. Robertson
- Florey Institute of Neurosciences & Mental Health, Parkville, VIC, Australia
| | - Simon M. Laws
- Centre for Precision Health, Edith Cowan University, Perth, WA, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| | - Paul Maruff
- Florey Institute of Neurosciences & Mental Health, Parkville, VIC, Australia
| | - Colin L. Masters
- Florey Institute of Neurosciences & Mental Health, Parkville, VIC, Australia
| | - Victor L. Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC, Australia
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christopher C. Rowe
- Florey Department of Neurosciences & Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC, Australia
- Florey Institute of Neurosciences & Mental Health, Parkville, VIC, Australia
| |
Collapse
|
30
|
Rane Levendovszky S. Cross-Sectional and Longitudinal Hippocampal Atrophy, Not Cortical Thinning, Occurs in Amyloid-Negative, p-Tau-Positive, Older Adults With Non-Amyloid Pathology and Mild Cognitive Impairment. FRONTIERS IN NEUROIMAGING 2022; 1:828767. [PMID: 37555137 PMCID: PMC10406207 DOI: 10.3389/fnimg.2022.828767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/11/2022] [Indexed: 08/10/2023]
Abstract
Introduction Alzheimer's disease (AD) is a degenerative disease characterized by pathological accumulation of amyloid and phosphorylated tau. Typically, the early stage of AD, also called mild cognitive impairment (MCI), shows amyloid pathology. A small but significant number of individuals with MCI do not exhibit amyloid pathology but have elevated phosphorylated tau levels (A-T+ MCI). We used CSF amyloid and phosphorylated tau to identify the individuals with A+T+ and A-T+ MCI as well as cognitively normal (A-T-) controls. To increase the sample size, we leveraged the Global Alzheimer's Association Interactive Network and identified 137 MCI+ and 61 A-T+ MCI participants. We compared baseline and longitudinal, hippocampal, and cortical atrophy between groups. Methods We applied ComBat harmonization to minimize site-related variability and used FreeSurfer for all measurements. Results Harmonization reduced unwanted variability in cortical thickness by 3.4% and in hippocampal volume measurement by 10.3%. Cross-sectionally, widespread cortical thinning with age was seen in the A+T+ and A-T+ MCI groups (p < 0.0005). A decrease in the hippocampal volume with age was faster in both groups (p < 0.05) than in the controls. Longitudinally also, hippocampal atrophy rates were significant (p < 0.05) when compared with the controls. No longitudinal cortical thinning was observed in A-T+ MCI group. Discussion A-T+ MCI participants showed similar baseline cortical thickness patterns with aging and longitudinal hippocampal atrophy rates as participants with A+T+ MCI, but did not show longitudinal cortical atrophy signature.
Collapse
Affiliation(s)
- Swati Rane Levendovszky
- Department of Radiology, School of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
31
|
Wu KY, Lin KJ, Chen CH, Liu CY, Wu YM, Chen CS, Yen TC, Hsiao IT. Decreased Cerebral Amyloid-β Depositions in Patients With a Lifetime History of Major Depression With Suspected Non-Alzheimer Pathophysiology. Front Aging Neurosci 2022; 14:857940. [PMID: 35721010 PMCID: PMC9204309 DOI: 10.3389/fnagi.2022.857940] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/11/2022] [Indexed: 11/19/2022] Open
Abstract
Cerebral amyloid-β (Aβ) depositions in depression in old age are controversial. A substantial proportion of individuals with late-life major depressive disorder (MDD) could be classified as having suspected non-Alzheimer’s disease pathophysiology (SNAP) by a negative test for the biomarker amyloid-β (Aβ−) but positive neurodegeneration (ND+). This study aimed to evaluate subthreshold Aβ loads in amyloid-negative MDD, particularly in SNAP MDD patients. This study included 46 amyloid-negative MDD patients: 23 SNAP (Aβ−/ND+) MDD and 23 Aβ−/ND− MDD, and 22 Aβ−/ND− control subjects. All subjects underwent 18F-florbetapir PET, FDG-PET, and MRI. Regions of interest (ROIs) and voxel-wise group comparisons were performed with adjustment for age, gender, and level of education. The SNAP MDD patients exhibited significantly deceased 18F-florbetapir uptakes in most cortical regions but not the parietal and precuneus cortex, as compared with the Aβ−/ND− MDD and control subjects (FDR correction, p < 0.05). No correlations of neuropsychological tests or depression characteristics with global cortical uptakes, but significant positive correlations between cognitive functions and adjusted hippocampal volumes among different groups were observed. The reduced Aβ depositions in the amyloid-negative MDD patients might be attributed mainly to the SNAP MDD patients. Our results indicated that meaningfully low amounts of subclinical Aβ might contain critical information on the non-amyloid-mediated pathogenesis.
Collapse
Affiliation(s)
- Kuan-Yi Wu
- Department of Psychiatry, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Kun-Ju Lin
- Department of Nuclear Medicine, Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
- Department of Medical Imaging and Radiological Sciences, College of Medicine and Healthy Aging Research Center, Chang Gung University, Taoyuan City, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
| | - Chia-Hsiang Chen
- Department of Psychiatry, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Chia-Yih Liu
- Department of Psychiatry, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Yi-Ming Wu
- Department of Radiology, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Cheng-Sheng Chen
- Department of Psychiatry, Kaohsiung Medical University Hospital, College of Medicine, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Tzu-Chen Yen
- Department of Nuclear Medicine, Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
- Department of Medical Imaging and Radiological Sciences, College of Medicine and Healthy Aging Research Center, Chang Gung University, Taoyuan City, Taiwan
| | - Ing-Tsung Hsiao
- Department of Nuclear Medicine, Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
- Department of Medical Imaging and Radiological Sciences, College of Medicine and Healthy Aging Research Center, Chang Gung University, Taoyuan City, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
- *Correspondence: Ing-Tsung Hsiao,
| |
Collapse
|
32
|
Giorgio J, Jagust WJ, Baker S, Landau SM, Tino P, Kourtzi Z. A robust and interpretable machine learning approach using multimodal biological data to predict future pathological tau accumulation. Nat Commun 2022; 13:1887. [PMID: 35393421 PMCID: PMC8989879 DOI: 10.1038/s41467-022-28795-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 02/11/2022] [Indexed: 01/21/2023] Open
Abstract
The early stages of Alzheimer's disease (AD) involve interactions between multiple pathophysiological processes. Although these processes are well studied, we still lack robust tools to predict individualised trajectories of disease progression. Here, we employ a robust and interpretable machine learning approach to combine multimodal biological data and predict future pathological tau accumulation. In particular, we use machine learning to quantify interactions between key pathological markers (β-amyloid, medial temporal lobe atrophy, tau and APOE 4) at mildly impaired and asymptomatic stages of AD. Using baseline non-tau markers we derive a prognostic index that: (a) stratifies patients based on future pathological tau accumulation, (b) predicts individualised regional future rate of tau accumulation, and (c) translates predictions from deep phenotyping patient cohorts to cognitively normal individuals. Our results propose a robust approach for fine scale stratification and prognostication with translation impact for clinical trial design targeting the earliest stages of AD.
Collapse
Affiliation(s)
- Joseph Giorgio
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
- Molecular Biophysics & Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Suzanne Baker
- Molecular Biophysics & Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Susan M Landau
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Peter Tino
- School of Computer Science, University of Birmingham, Birmingham, UK
| | - Zoe Kourtzi
- Department of Psychology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
33
|
Casanova R, Hsu FC, Barnard RT, Anderson AM, Talluri R, Whitlow CT, Hughes TM, Griswold M, Hayden KM, Gottesman RF, Wagenknecht LE. Comparing data-driven and hypothesis-driven MRI-based predictors of cognitive impairment in individuals from the Atherosclerosis Risk in Communities (ARIC) study. Alzheimers Dement 2022; 18:561-571. [PMID: 34310039 PMCID: PMC8789939 DOI: 10.1002/alz.12427] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 06/15/2021] [Accepted: 06/15/2021] [Indexed: 01/10/2023]
Abstract
INTRODUCTION A data-driven index of dementia risk based on magnetic resonance imaging (MRI), the Alzheimer's Disease Pattern Similarity (AD-PS) score, was estimated for participants in the Atherosclerosis Risk in Communities (ARIC) study. METHODS AD-PS scores were generated for 839 cognitively non-impaired individuals with a mean follow-up of 4.86 years. The scores and a hypothesis-driven volumetric measure based on several brain regions susceptible to AD were compared as predictors of incident cognitive impairment in different settings. RESULTS Logistic regression analyses suggest the data-driven AD-PS scores to be more predictive of incident cognitive impairment than its counterpart. Both biomarkers were more predictive of incident cognitive impairment in participants who were White, female, and apolipoprotein E gene (APOE) ε4 carriers. Random forest analyses including predictors from different domains ranked the AD-PS scores as the most relevant MRI predictor of cognitive impairment. CONCLUSIONS Overall, the AD-PS scores were the stronger MRI-derived predictors of incident cognitive impairment in cognitively non-impaired individuals.
Collapse
Affiliation(s)
- Ramon Casanova
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem
| | - Ryan T. Barnard
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem
| | - Andrea M. Anderson
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem
| | - Rajesh Talluri
- University of Mississippi Medical Center, Jackson, MS, USA
| | | | - Timothy M. Hughes
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | - Kathleen M. Hayden
- Department of Social Sciences and Health Policy, Wake Forest School of Medicine, Winston-Salem
| | | | - Lynne E. Wagenknecht
- Divison of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | |
Collapse
|
34
|
Li TR, Yang Q, Hu X, Han Y. Biomarkers and Tools for Predicting Alzheimer's Disease in the Preclinical Stage. Curr Neuropharmacol 2022; 20:713-737. [PMID: 34030620 PMCID: PMC9878962 DOI: 10.2174/1570159x19666210524153901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 04/27/2021] [Accepted: 05/08/2021] [Indexed: 11/22/2022] Open
Abstract
Alzheimer's disease (AD) is the only leading cause of death for which no disease-modifying therapy is currently available. Over the past decade, a string of disappointing clinical trial results has forced us to shift our focus to the preclinical stage of AD, which represents the most promising therapeutic window. However, the accurate diagnosis of preclinical AD requires the presence of brain β- amyloid deposition determined by cerebrospinal fluid or amyloid-positron emission tomography, significantly limiting routine screening and diagnosis in non-tertiary hospital settings. Thus, an easily accessible marker or tool with high sensitivity and specificity is highly needed. Recently, it has been discovered that individuals in the late stage of preclinical AD may not be truly "asymptomatic" in that they may have already developed subtle or subjective cognitive decline. In addition, advances in bloodderived biomarker studies have also allowed the detection of pathologic changes in preclinical AD. Exosomes, as cell-to-cell communication messengers, can reflect the functional changes of their source cell. Methodological advances have made it possible to extract brain-derived exosomes from peripheral blood, making exosomes an emerging biomarker carrier and liquid biopsy tool for preclinical AD. The eye and its associated structures have rich sensory-motor innervation. In this regard, studies have indicated that they may also provide reliable markers. Here, our report covers the current state of knowledge of neuropsychological and eye tests as screening tools for preclinical AD and assesses the value of blood and brain-derived exosomes as carriers of biomarkers in conjunction with the current diagnostic paradigm.
Collapse
Affiliation(s)
- Tao-Ran Li
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Qin Yang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Xiaochen Hu
- Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, 50924, Germany
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China;,Center of Alzheimer’s Disease, Beijing Institute for Brain Disorders, Beijing, 100053, China;,National Clinical Research Center for Geriatric Disorders, Beijing, 100053, China;,School of Biomedical Engineering, Hainan University, Haikou, 570228, China;,Address correspondence to this author at the Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China; Tel: +86 13621011941; E-mail:
| |
Collapse
|
35
|
Woodfield A, Porter T, Gilani I, Noordin S, Li QX, Collins S, Martins RN, Maruff P, Masters CL, Rowe CC, Villemagne VL, Dore V, Newsholme P, Laws SM, Verdile G. Insulin resistance, cognition and Alzheimer's disease biomarkers: Evidence that CSF Aβ42 moderates the association between insulin resistance and increased CSF tau levels. Neurobiol Aging 2022; 114:38-48. [DOI: 10.1016/j.neurobiolaging.2022.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/09/2022] [Accepted: 03/07/2022] [Indexed: 12/16/2022]
|
36
|
Jack CR, Therneau TM, Lundt ES, Wiste HJ, Mielke MM, Knopman DS, Graff-Radford J, Lowe VJ, Vemuri P, Schwarz CG, Senjem ML, Gunter JL, Petersen RC. Long-term associations between amyloid positron emission tomography, sex, apolipoprotein E and incident dementia and mortality among individuals without dementia: hazard ratios and absolute risk. Brain Commun 2022; 4:fcac017. [PMID: 35310829 PMCID: PMC8924651 DOI: 10.1093/braincomms/fcac017] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/08/2021] [Accepted: 01/31/2022] [Indexed: 11/14/2022] Open
Abstract
Dementia and mortality rates rise inexorably with age and consequently interact. However, because of the major logistical difficulties in accounting for both outcomes in a defined population, very little work has examined how risk factors and biomarkers for incident dementia are influenced by competing mortality. The objective of this study was to examine long-term associations between amyloid PET, APOE ɛ4, sex, education and cardiovascular/metabolic conditions, and hazard and absolute risk of dementia and mortality in individuals without dementia at enrolment. Participants were enrolled in the Mayo Clinic Study of Aging, a population-based study of cognitive ageing in Olmsted County, MN, USA. All were without dementia and were age 55-92 years at enrolment and were followed longitudinally. Predictor variables were amyloid PET, APOE ɛ4 status, sex, education, cardiovascular/metabolic conditions and age. The main outcomes were incident dementia and mortality. Multivariable, multi-state models were used to estimate mortality and incident dementia rates and absolute risk of dementia and mortality by predictor variable group. Of the 4984 participants in the study, 4336 (87%) were cognitively unimpaired and 648 (13%) had mild cognitive impairment at enrolment. The median age at enrolment was 75 years; 2463 (49%) were women. The median follow-up time was 9.4 years (7.5 years after PET). High versus normal amyloid (hazard ratio 2.11, 95% confidence interval 1.43-2.79), APOE ɛ4 (women: hazard ratio 2.24, 95% confidence interval 1.80-2.77; men: hazard ratio 1.37, 95% confidence interval 1.09-1.71), older age and two additional cardiovascular/metabolic conditions (hazard ratio 1.37, 95% confidence interval 1.22-1.53) were associated with the increased hazard of dementia (all P < 0.001). Among APOE ɛ4 carriers with elevated amyloid, remaining lifetime risk of dementia at age 65 years was greater in women [74% (95% confidence interval 65-84%) high and 58% (95% confidence interval 52-65%) moderate amyloid], than men [62% (95% confidence interval 52-73%) high and 44% (95% confidence interval 35-53%) moderate amyloid]. Overall, the hazard and absolute risk of dementia varied considerably by predictor group. The absolute risk of dementia associated with predictors characteristic of Alzheimer's disease was greater in women than men while at the same time the combination of APOE ɛ4 non-carrier with normal amyloid was more protective in women than men. This set of findings may be attributed in part to different biological effects and in part to lower mortality rates in women.
Collapse
Affiliation(s)
| | - Terry M. Therneau
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Emily S. Lundt
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Heather J. Wiste
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Michelle M. Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Val J. Lowe
- Department of Nuclear Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | |
Collapse
|
37
|
Mortamais M, Laure-Anne G, Balem M, Bars EL, de Champfleur NM, Bouyahia A, Chupin M, Perus L, Fisher C, Vellas B, Andrieu S, Mangin JF, Berr C, Gabelle A. Sulcal morphology as cognitive decline predictor in older adults with memory complaints. Neurobiol Aging 2022; 113:84-94. [DOI: 10.1016/j.neurobiolaging.2022.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/08/2021] [Accepted: 02/08/2022] [Indexed: 11/16/2022]
|
38
|
Gagliardi G, Vannini P. Episodic Memory Impairment Mediates the Loss of Awareness in Mild Cognitive Impairment. Front Aging Neurosci 2022; 13:802501. [PMID: 35126092 PMCID: PMC8814670 DOI: 10.3389/fnagi.2021.802501] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/30/2021] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION Loss of awareness is a common symptom in Alzheimer's Disease (AD) and responsible for a significant loss of functional abilities. The mechanisms underlying loss of awareness in AD is unknown, although previous findings have implicated dysfunction of primary executive functioning (EF) or episodic memory (EM) to be the cause. Therefore, our main study objective was to explore the involvement of EF and EM dysfunction in amyloid-related loss of awareness across the clinical spectrum of AD. METHODS A total of 895 participants (362 clinically normal [CN], 422 people with mild cognitive impairment [MCI] and 111 with dementia) from the Alzheimer's Disease Neuroimaging Initiative were used for the analyses. A sub-analysis was performed in 202 participants who progressed in their clinical diagnosis from CN to MCI or MCI to dementia as well as dementia patients. Mediation models were used in each clinical group with awareness (assessed with the Everyday Cognitive function questionnaire) as a dependent variable to determine whether EF and/or EM would mediate the effect of amyloid on awareness. We also ran these analyses with subjective and informant complaints as dependent variables. Direct correlations between all variables were also performed. RESULTS We found evidence for a decline in awareness across the groups, with increased awareness observed in the CN group and decreased awareness observed in the MCI and dementia groups. Our results showed that EM, and not EF, partially mediated the relationship between amyloid and awareness such that greater amyloid and lower EM performance was associated with lower awareness. When analyzing each group separately, this finding was only observed in the MCI group and in the group containing progressors and dementia patients. When repeating the analyses for subjective and informant complaints separately, the results were replicated only for the informant's complaints. DISCUSSION Our results demonstrate that decline in EM and, to a lesser degree, EF, mediate the effect of amyloid on awareness. In line with previous studies demonstrating the development of anosognosia in the prodromal stage, our findings suggest that decreased awareness is the result of an inability for the participant to update his/her insight into his/her cognitive performance (i.e., demonstrating a petrified self).
Collapse
Affiliation(s)
- Geoffroy Gagliardi
- Neurology, Brigham and Women's Hospital, Boston, MA, United States
- Neurology, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Cambridge, MA, United States
| | - Patrizia Vannini
- Neurology, Brigham and Women's Hospital, Boston, MA, United States
- Neurology, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Cambridge, MA, United States
| |
Collapse
|
39
|
de Mendonça Lima CA. Towards a WPA Position Document on the Human Rights of Older Adults with Mental Health Conditions: К документу с изложением позиции Всемирной психиатрической ассоциации по вопросу соблюдения прав пожилых людей с психическими расстройствами. CONSORTIUM PSYCHIATRICUM 2022; 3:16-21. [PMID: 39045355 PMCID: PMC11262087 DOI: 10.17816/cp150] [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: 02/01/2022] [Accepted: 02/12/2022] [Indexed: 11/08/2022] Open
Abstract
The increasing number of older adults in countries across the world is a huge challenge to those that are in charge of promoting, protecting, and implementing their human rights. This task is particularly difficult in the absence of a strong international framework addressing the principles required to guide the actions to combat all human rights violations. The existence of such a specific framework for older adults with mental health conditions is justified in view of the particular vulnerability of this section of the population by virtue of societal ageism, stigmatization, exclusion, as well as the disability and dependency which mental health conditions in old age may confer. The present article is a development of a previous statement by the International Psychogeriatric Association and the World Psychiatric Association Section of Old Age Psychiatry. As there is a call to all organizations to support efforts to combat Human Rights violations among older adults, a text will be submitted to the Executive Committee of the World Psychiatric Association to approve an official position statement on Human Rights of Older Persons with Mental Health Conditions.
Collapse
|
40
|
Gavrilova S. The Evolution of Diagnostic Boundaries of Alzheimer's Disease and Novel Therapeutic Options: Эволюция диагностических границ болезни Альцгеймера и новые терапевтические возможности. CONSORTIUM PSYCHIATRICUM 2022; 3:8-15. [PMID: 39045353 PMCID: PMC11262098 DOI: 10.17816/cp152] [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: 02/04/2022] [Accepted: 02/22/2022] [Indexed: 11/08/2022] Open
Abstract
Over the past three decades, the definition and diagnostic boundaries of Alzheimer's disease (AD) have been repeatedly revised due to significant progress in understanding of the pathogenesis of neurodegeneration associated with Alzheimer's disease and in the development of high-tech diagnostic methods. The current approach to AD diagnostics relies on the detection of biomarkers that reflect two main neuropathological processes involved in the primary neurodegeneration that underlies AD: abnormal amyloidogenesis, and neuronal degeneration. The currently available diagnostic tools are limited to the detection of cerebrospinal biomarkers and/or assessment of the abnormal amyloid and tau protein burden in the brain via amyloid and tau positron emission tomography (PET) ligands. Practical implementation (mostly in the research field) of the biological model of AD diagnosis has led to a significant expansion of its diagnostic boundaries with the inclusion of predementia AD stages: asymptomatic and symptomatic, the latter is clinically corresponding to amnestic mild cognitive impairment (aMCI-amnestic mild cognitive impairment). On the one hand, this approach significantly expands the possibilities to study and use preventive technologies aiming to avert or delay the progression of predementia cognitive impairment to dementia but, on the other, it is associated with a number of negative implications from both the clinical and ethical points of view. A significant limitation of purely biological diagnosis of AD based on biomarker levels is due to the low prognostic value of biomarkers, which can cause diagnostic confusion in certain circumstances. Moreover, since the future evolution of the asymptomatic stage is not yet clear and there are still no reliable ways to prevent the cognitive and behavioral symptoms associated with AD, disclosure of stressful information about this "terrifying" diagnosis to patients can cause irreversible damage by triggering depressive disorder, which is a risk factor of AD itself. The current knowledge about AD prognosis in amyloid-positive cognitively unimpaired patients is insufficient.The most adequate approach to early AD diagnostics appears to be the clinical and biological model, as recommended by the International Working Group (IWG 2021), which requires a combination of the clinical AD phenotype and the detection of biomarkers specific to this disease. The article discusses the potential directions for the development of biological diagnostic methods, including those based on the so-called peripheral (serum) biomarker technologies and promising directions for the development of biological methods of secondary AD prevention.
Collapse
|
41
|
Kim YK. Recent Updates on PET Imaging in Neurodegenerative Diseases. JOURNAL OF THE KOREAN SOCIETY OF RADIOLOGY 2022; 83:453-472. [PMID: 36238518 PMCID: PMC9514517 DOI: 10.3348/jksr.2022.0052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/08/2022] [Accepted: 05/16/2022] [Indexed: 11/23/2022]
Abstract
양전자방출단층촬영(PET)을 이용한 단백질병리의 생체영상기술은 퇴행성 치매의 질병 기전을 이해하는데 필요한 정보를 제공할 뿐 아니라, 질병의 조기 발견과 치료법 개발에서 중요한 역할을 수행하고 있다. 베타아밀로이드와 타우 PET 영상은 인체 뇌병리에 기반한 알츠하이머병 연속체에 대한 진단 바이오마커로 확립되어 조기진단과 감별진단을 용이하게 하고, 질병 예후를 예측하고 있다. 또한, 치매치료제 개발에서 예후 및 대리 바이오마커로의 역할이 커지고 있다. 이 종설에서는 치매를 유발하는 알츠하이머병 및 기타 퇴행성 뇌질환에서 베타아밀로이드와 타우 단백질의 뇌축적을 영상화하는 PET의 최근 임상적 적용과 최근 동향을 살펴보고, 잠재적 유용성을 소개하고자 한다.
Collapse
Affiliation(s)
- Yu Kyeong Kim
- Department of Nuclear Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| |
Collapse
|
42
|
Sarant JZ, Harris DC, Busby PA, Fowler C, Fripp J, Masters CL, Maruff P. No Influence of Age-Related Hearing Loss on Brain Amyloid-β. J Alzheimers Dis 2022; 85:359-367. [PMID: 34806606 PMCID: PMC8842788 DOI: 10.3233/jad-215121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Hearing loss is independently associated with a faster rate of cognitive decline in older adults and has been identified as a modifiable risk factor for dementia. The mechanism for this association is unknown, and there has been limited exploration of potential casual pathology. OBJECTIVE Our objective was to investigate whether there was an association between degree of audiometrically measured hearing loss (HL) and brain amyloid-β (Aβ) in a pre-clinical sample. METHODS Participants of the Australian Imaging and Biomarker Longitudinal Study (AIBL; n = 143) underwent positron emission tomography (PET) imaging and objective measurement of hearing thresholds within 5 years of imaging, as well as cognitive assessment within 2 years of imaging in this observational cohort study. RESULTS With one exception, study participants who had cognitive assessments within 2 years of their PET imaging (n = 113) were classified as having normal cognition. There was no association between cognitive scores and degree of hearing loss, or between cognitive scores and Aβ load. No association between HL and Aβ load was found once age was controlled for. As previously reported, positive Apolipoprotein E4 (APOE4) carrier status increased the risk of being Aβ positive (p = 0.002). CONCLUSION Degree of HL was not associated with positive Aβ status.
Collapse
Affiliation(s)
| | | | | | | | - Jurgen Fripp
- Commonwealth Scientific and Industrial Research Organization, Queensland, Australia
| | | | | |
Collapse
|
43
|
Gavrilova S. Evolution of the diagnostic frontiers of Alzheimer’s disease and new therapeutic possibilities. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:38-44. [DOI: 10.17116/jnevro202212211238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
44
|
Characteristics of Bipolar Patients with Cognitive Impairment of Suspected Neurodegenerative Origin: A Multicenter Cohort. J Pers Med 2021; 11:jpm11111183. [PMID: 34834535 PMCID: PMC8620397 DOI: 10.3390/jpm11111183] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/29/2021] [Accepted: 11/09/2021] [Indexed: 11/17/2022] Open
Abstract
Bipolar disorder is associated with an increased risk of dementia with aging. Little is known regarding this association, limiting appropriate diagnosis and management. We aimed to describe the characteristics of bipolar patients with late cognitive impairment for whom the hypothesis of an underlying neurodegenerative disease had been raised. We performed a retrospective multicenter study, recruiting bipolar patients over 50 years old from five French tertiary memory centers who had undergone cerebrospinal fluid (CSF) biomarker assessment for Alzheimer’s disease (AD). Clinical, neuropsychological, and paraclinical characteristics were analyzed and 78 patients were included. The mean age at the onset of cognitive impairment was 62.4 years (±9.2). The mean MMSE score was 22.8 (±4.5), the mean FAB was 11.7 (±3.9), and the mean FCRST was 15.8 (±7.4)/36.8 (±9.7) (free/total recall). A total of 48.6% of the patients displayed cognitive fluctuations, and 38.2% showed cognitive improvement during follow-ups; and 56.3% of the patients showed Parkinsonism, of which 12.7% had never received antipsychotics. Among patients who underwent DAT-scans, 35.3% displayed dopaminergic denervation; 10.3% of patients had CSF AD biological signature (“A+ T+” profile), while 56.4% had other abnormal CSF profiles. Thus, clinical presentation was dominated by executive dysfunction, episodic memory impairment, fluctuating cognition, and a high frequency of Parkinsonism. Specifically, high frequency of delusional episodes suggests limited tolerance of psychotropic drugs. Most patients had abnormal CSF biomarker profiles, but only a minority displayed AD’s specific biomarker signature. Therefore, while our results unveil shared common neurocognitive features in bipolar patients with cognitive impairment of suspected neurodegenerative origin they suggest a participation of various underlying pathologies rather than a common degenerative mechanism in the pathophysiology of this condition.
Collapse
|
45
|
Rizzi L, Balthazar MLF. Mini-review: The suspected non-Alzheimer's disease pathophysiology. Neurosci Lett 2021; 764:136208. [PMID: 34478819 DOI: 10.1016/j.neulet.2021.136208] [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: 07/18/2021] [Revised: 08/13/2021] [Accepted: 08/24/2021] [Indexed: 11/17/2022]
Abstract
Suspected non-Alzheimer's disease pathophysiology (SNAP) is a biomarker-based concept that underlying etiology has not been completely understood. Refers to a group of individuals that are negative for amyloid biomarkers and positive for p-Tau and/or neurodegeneration. SNAP causes great research interest because it is not clear if they have a different biological basis from Alzheimer's disease (AD), or are in an early stage of AD itself. The pathological processes behind SNAP need to be clarified. This mini-review aims to summarize the main characteristics of SNAP, besides reporting challenges and promising biomarkers related to the concept.
Collapse
Affiliation(s)
- Liara Rizzi
- Department of Neurology, University of Campinas (UNICAMP), Campinas, SP, Brazil.
| | | |
Collapse
|
46
|
Yoon B, Guo T, Provost K, Korman D, Ward TJ, Landau SM, Jagust WJ. Abnormal tau in amyloid PET negative individuals. Neurobiol Aging 2021; 109:125-134. [PMID: 34715443 DOI: 10.1016/j.neurobiolaging.2021.09.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 09/03/2021] [Accepted: 09/22/2021] [Indexed: 12/14/2022]
Abstract
We examined the characteristics of individuals with biomarker evidence of tauopathy but without β-amyloid (Aβ) (A-T+) in relation to individuals with (A+T+) and without (A-T-) evidence of Alzheimer's disease (AD). We included 561 participants with Aβ and tau PET from the Alzheimer's Disease Neuroimaging Initiative (ADNI). We compared A-T- (n = 316), A-T+ (n = 63), and A+T+ (n = 182) individuals on demographics, amyloid, tau, hippocampal volumes, and cognition. A-T+ individuals were low on apolipoprotein E ɛ4 prevalence (17%) and had no evidence of subtly elevated brain Aβ within the negative range. The severity of tau deposition, hippocampal atrophy, and cognitive dysfunction in the A-T+ group was intermediate between A-T- and A+T+ (all p < 0.001). Tau uptake patterns in A-T+ individuals were heterogeneous, but approximately 29% showed tau deposition in the medial temporal lobe only, consistent with primary age-related tauopathy and an additional 32% showed a pattern consistent with AD. A-T+ individuals also share other features that are characteristic of AD such as cognitive impairment and neurodegeneration, but this group is heterogeneous and likely reflects more than one disorder.
Collapse
Affiliation(s)
- Bora Yoon
- Department of Neurology, Konyang University Hospital, Konyang University, College of Medicine, Daejeon, Korea.
| | - Tengfei Guo
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA; Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Karine Provost
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Deniz Korman
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Tyler J Ward
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Susan M Landau
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA; Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| |
Collapse
|
47
|
Darmanthé N, Tabatabaei-Jafari H, Cherbuin N. Combination of Plasma Neurofilament Light Chain and Mini-Mental State Examination Score Predicts Progression from Mild Cognitive Impairment to Alzheimer's Disease within 5 Years. J Alzheimers Dis 2021; 82:951-964. [PMID: 34120902 DOI: 10.3233/jad-210092] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Individuals with mild cognitive impairment (MCI) are at high risk of progression to Alzheimer's disease (AD) dementia, but some remain stable. There is a need to identify those at higher risk of progression to improve patient management and outcomes. OBJECTIVE To evaluate the trajectory of plasma neurofilament light chain (pNFL) prior to progression from MCI to AD dementia, the performance of pNFL, in combination with the Mini-Mental State Examination (MMSE), as a predictor of progression from MCI to AD dementia and to inform clinicians on the use of pNFL as a predictive biomarker. METHODS Participants (n = 440) with MCI and longitudinal follow-up (mean = 4.2 years) from the AD Neuroimaging Initiative dataset were included. pNFL as a marker for neurodegeneration and the MMSE as a cognitive measure were investigated as simple/practical predictors of progression. The risk of progressing from MCI to AD dementia associated with pNFL and MMSE scores was assessed using Cox and logistic regression models. RESULTS The current risk of progression to AD dementia was 37%higher in individuals with high pNFL (> 56 ng/L) compared to those with average pNFL (≤40 ng/L). A combination of baseline pNFL and MMSE could differentiate those who progressed within 5 years (AUC = 0.75) from stable individuals. Including change in MMSE over 6-12 months further improved the model (AUC = 0.84). CONCLUSION Our findings reveal that combining pNFL with a simple dementia screener (MMSE) can reliably predict whether a person with MCI is likely to progress to AD dementia within 5 years.
Collapse
Affiliation(s)
- Nicolas Darmanthé
- ANU Medical School, Australian National University, Canberra, Australia
| | - Hossein Tabatabaei-Jafari
- Centre for Research on Ageing, Health and Wellbeing, Australian National University, Canberra, Australia
| | - Nicolas Cherbuin
- Centre for Research on Ageing, Health and Wellbeing, Australian National University, Canberra, Australia
| | | |
Collapse
|
48
|
Molinder A, Ziegelitz D, Maier SE, Eckerström C. Validity and reliability of the medial temporal lobe atrophy scale in a memory clinic population. BMC Neurol 2021; 21:289. [PMID: 34301202 PMCID: PMC8305846 DOI: 10.1186/s12883-021-02325-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 07/12/2021] [Indexed: 11/30/2022] Open
Abstract
Background Visual rating of medial temporal lobe atrophy (MTA) is often performed in conjunction with dementia workup. Most prior studies involved patients with known or probable Alzheimer’s disease (AD). This study investigated the validity and reliability of MTA in a memory clinic population. Methods MTA was rated in 752 MRI examinations, of which 105 were performed in cognitively healthy participants (CH), 184 in participants with subjective cognitive impairment, 249 in subjects with mild cognitive impairment, and 214 in patients with dementia, including AD, subcortical vascular dementia and mixed dementia. Hippocampal volumes, measured manually or using FreeSurfer, were available in the majority of cases. Intra- and interrater reliability was tested using Cohen’s weighted kappa. Correlation between MTA and quantitative hippocampal measurements was ascertained with Spearman’s rank correlation coefficient. Moreover, diagnostic ability of MTA was assessed with receiver operating characteristic (ROC) analysis and suitable, age-dependent MTA thresholds were determined. Results Rater agreement was moderate to substantial. MTA correlation with quantitative volumetric methods ranged from -0.20 (p< 0.05) to -0.68 (p < 0.001) depending on the quantitative method used. Both MTA and FreeSurfer are able to distinguish dementia subgroups from CH. Suggested age-dependent MTA thresholds are 1 for the age group below 75 years and 1.5 for the age group 75 years and older. Conclusions MTA can be considered a valid marker of medial temporal lobe atrophy and may thus be valuable in the assessment of patients with cognitive impairment, even in a heterogeneous patient population.
Collapse
Affiliation(s)
- Anna Molinder
- Department of Radiology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden. .,Neuroradiology, Sahlgrenska sjukhuset, Blå stråket 5, Gothenburg, 413 46, Sweden.
| | - Doerthe Ziegelitz
- Department of Radiology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Stephan E Maier
- Department of Radiology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Carl Eckerström
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Immunology and Transfusion Medicine, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
49
|
Alongi P, Chiaravalloti A, Berti V, Vellani C, Trifirò G, Puccini G, Carli G, Chincarini A, Morbelli S, Perani D, Sestini S. Amyloid PET in the diagnostic workup of neurodegenerative disease. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-021-00428-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
50
|
Zhuo J, Zhang Y, Liu Y, Liu B, Zhou X, Bartlett PF, Jiang T. New Trajectory of Clinical and Biomarker Changes in Sporadic Alzheimer's Disease. Cereb Cortex 2021; 31:3363-3373. [PMID: 33690839 DOI: 10.1093/cercor/bhab017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/08/2020] [Accepted: 01/18/2021] [Indexed: 12/20/2022] Open
Abstract
Identifying dynamic changes in biomarkers and clinical profiles is essential for understanding the progression of Alzheimer's disease (AD). The relevant studies have primarily relied on patients with autosomal dominant AD; however, relevant studies in sporadic AD are poorly understood. Here, we analyzed longitudinal data from 665 participants (mean follow-up 4.90 ± 2.83 years). By aligning normal cognition (CN) baseline with a clinical diagnosis of mild cognitive impairment (MCI) or AD, we studied the progression of AD using a linear mixed model to estimate the clinical and biomarker changes from stable CN to MCI to AD. The results showed that the trajectory of hippocampal volume and fluorodeoxyglucose (FDG) was consistent with the clinical measures in that they did not follow a hypothetical sigmoid curve but rather showed a slow change in the initial stage and accelerated changes in the later stage from MCI conversion to AD. Dramatic hippocampal atrophy and the ADAS13 increase were, respectively, 2.5 and 1 years earlier than the MCI onset. Besides, cognitively normal people with elevated and normal amyloid showed no significant differences in clinical measures, hippocampal volume, or FDG. These results reveal that pre-MCI to pre-AD may be a better time window for future clinical trial design.
Collapse
Affiliation(s)
- Junjie Zhuo
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,The Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia.,School of Biomedical Engineering, Hainan University, Haikou 570228, China
| | - Yuanchao Zhang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 625014, China
| | - Yong Liu
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,CAS Center for Excellence in Brain Science, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Bing Liu
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,CAS Center for Excellence in Brain Science, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaoqing Zhou
- The Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
| | - Perry F Bartlett
- The Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
| | - Tianzi Jiang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,The Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia.,University of Chinese Academy of Sciences, Beijing 100049, China.,CAS Center for Excellence in Brain Science, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 625014, China
| | | |
Collapse
|