1
|
Yang B, Earnest T, Kumar S, Kothapalli D, Benzinger T, Gordon B, Sotiras A. Evaluation of ComBat harmonization for reducing across-tracer differences in regional amyloid PET analyses. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.14.24308952. [PMID: 38947044 PMCID: PMC11213066 DOI: 10.1101/2024.06.14.24308952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Introduction Differences in amyloid positron emission tomography (PET) radiotracer pharmacokinetics and binding properties lead to discrepancies in amyloid-β uptake estimates. Harmonization of tracer-specific biases is crucial for optimal performance of downstream tasks. Here, we investigated the efficacy of ComBat, a data-driven harmonization model, for reducing tracer-specific biases in regional amyloid PET measurements from [18F]-florbetapir (FBP) and [11C]-Pittsburgh Compound-B (PiB). Methods One-hundred-thirteen head-to-head FBP-PiB scan pairs, scanned from the same subject within ninety days, were selected from the Open Access Series of Imaging Studies 3 (OASIS-3) dataset. The Centiloid scale, ComBat with no covariates, ComBat with biological covariates, and GAM-ComBat with biological covariates were used to harmonize both global and regional amyloid standardized uptake value ratios (SUVR). Variants of ComBat, including longitudinal ComBat and PEACE, were also tested. Intraclass correlation coefficient (ICC) and mean absolute error (MAE) were computed to measure the absolute agreement between tracers. Additionally, longitudinal amyloid SUVRs from an anti-amyloid drug trial were simulated using linear mixed effects modeling. Differences in rates-of-change between simulated treatment and placebo groups were tested, and change in statistical power/Type-I error after harmonization was quantified. Results In the head-to-head tracer comparison, ComBat with no covariates was the best at increasing ICC and decreasing MAE of both global summary and regional amyloid PET SUVRs between scan pairs of the same group of subjects. In the clinical trial simulation, harmonization with both Centiloid and ComBat increased statistical power of detecting true rate-of-change differences between groups and decreased false discovery rate in the absence of a treatment effect. The greatest benefit of harmonization was observed when groups exhibited differing FBP-to-PiB proportions. Conclusion ComBat outperformed the Centiloid scale in harmonizing both global and regional amyloid estimates. Additionally, ComBat improved the detection of rate-of-change differences between clinical trial groups. Our findings suggest that ComBat is a viable alternative to Centiloid for harmonizing regional amyloid PET analyses.
Collapse
Affiliation(s)
- Braden Yang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA 63110
| | - Tom Earnest
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA 63110
| | - Sayantan Kumar
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA 63110
| | - Deydeep Kothapalli
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA 63110
| | - Tammie Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA 63110
| | - Brian Gordon
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA 63110
| | - Aristeidis Sotiras
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA 63110
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA 63110
| |
Collapse
|
2
|
Wisch JK, Gordon BA, Barthélemy NR, Horie K, Henson RL, He Y, Flores S, Benzinger TLS, Morris JC, Bateman RJ, Ances BM, Schindler SE. Predicting continuous amyloid PET values with CSF tau phosphorylation occupancies. Alzheimers Dement 2024; 20:6365-6373. [PMID: 39041391 PMCID: PMC11497729 DOI: 10.1002/alz.14132] [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: 04/24/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Cerebrospinal fluid (CSF) tau phosphorylation at multiple sites is associated with cortical amyloid and other pathologic changes in Alzheimer's disease. These relationships can be non-linear. We used an artificial neural network to assess the ability of 10 different CSF tau phosphorylation sites to predict continuous amyloid positron emission tomography (PET) values. METHODS CSF tau phosphorylation occupancies at 10 sites (including pT181/T181, pT217/T217, pT231/T231 and pT205/T205) were measured by mass spectrometry in 346 individuals (57 cognitively impaired, 289 cognitively unimpaired). We generated synthetic amyloid PET scans using biomarkers and evaluated their performance. RESULTS Concentration of CSF pT217/T217 had low predictive error (average error: 13%), but also a low predictive range (ceiling 63 Centiloids). CSF pT231/T231 has slightly higher error (average error: 19%) but predicted through a greater range (87 Centiloids). DISCUSSION Tradeoffs exist in biomarker selection. Some phosphorylation sites offer greater concordance with amyloid PET at lower levels, while others perform better over a greater range. HIGHLIGHTS Novel pTau isoforms can predict cortical amyloid burden. pT217/T217 accurately predicts cortical amyloid burden in low-amyloid individuals. Traditional CSF biomarkers correspond with higher levels of amyloid.
Collapse
Affiliation(s)
- Julie K. Wisch
- Department of NeurologyWashington University in St. LouisSt. LouisMissouriUSA
| | - Brian A. Gordon
- Department of RadiologyWashington University in St. LouisSt. LouisMissouriUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt LouisMissouriUSA
| | - Nicolas R. Barthélemy
- Department of NeurologyWashington University in St. LouisSt. LouisMissouriUSA
- SILQ Center for Neurodegenerative BiologySt. LouisMissouriUSA
| | - Kanta Horie
- Department of NeurologyWashington University in St. LouisSt. LouisMissouriUSA
- SILQ Center for Neurodegenerative BiologySt. LouisMissouriUSA
| | - Rachel L. Henson
- Hope CenterWashington University in Saint LouisSt. LouisMissouriUSA
| | - Yingxin He
- Department of NeurologyWashington University in St. LouisSt. LouisMissouriUSA
- SILQ Center for Neurodegenerative BiologySt. LouisMissouriUSA
| | - Shaney Flores
- Department of RadiologyWashington University in St. LouisSt. LouisMissouriUSA
| | - Tammie L. S. Benzinger
- Department of RadiologyWashington University in St. LouisSt. LouisMissouriUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt LouisMissouriUSA
| | - John C. Morris
- Department of NeurologyWashington University in St. LouisSt. LouisMissouriUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt LouisMissouriUSA
| | - Randall J. Bateman
- Department of NeurologyWashington University in St. LouisSt. LouisMissouriUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt LouisMissouriUSA
- SILQ Center for Neurodegenerative BiologySt. LouisMissouriUSA
- Hope CenterWashington University in Saint LouisSt. LouisMissouriUSA
| | - Beau M. Ances
- Department of NeurologyWashington University in St. LouisSt. LouisMissouriUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt LouisMissouriUSA
| | - Suzanne E. Schindler
- Department of NeurologyWashington University in St. LouisSt. LouisMissouriUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt LouisMissouriUSA
- Hope CenterWashington University in Saint LouisSt. LouisMissouriUSA
| |
Collapse
|
3
|
Daniels AJ, McDade E, Llibre-Guerra JJ, Xiong C, Perrin RJ, Ibanez L, Supnet-Bell C, Cruchaga C, Goate A, Renton AE, Benzinger TL, Gordon BA, Hassenstab J, Karch C, Popp B, Levey A, Morris J, Buckles V, Allegri RF, Chrem P, Berman SB, Chhatwal JP, Farlow MR, Fox NC, Day GS, Ikeuchi T, Jucker M, Lee JH, Levin J, Lopera F, Takada L, Sosa AL, Martins R, Mori H, Noble JM, Salloway S, Huey E, Rosa-Neto P, Sánchez-Valle R, Schofield PR, Roh JH, Bateman RJ. 15 Years of Longitudinal Genetic, Clinical, Cognitive, Imaging, and Biochemical Measures in DIAN. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.08.24311689. [PMID: 39148846 PMCID: PMC11326320 DOI: 10.1101/2024.08.08.24311689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
This manuscript describes and summarizes the Dominantly Inherited Alzheimer Network Observational Study (DIAN Obs), highlighting the wealth of longitudinal data, samples, and results from this human cohort study of brain aging and a rare monogenic form of Alzheimer's disease (AD). DIAN Obs is an international collaborative longitudinal study initiated in 2008 with support from the National Institute on Aging (NIA), designed to obtain comprehensive and uniform data on brain biology and function in individuals at risk for autosomal dominant AD (ADAD). ADAD gene mutations in the amyloid protein precursor (APP), presenilin 1 (PSEN1), or presenilin 2 (PSEN2) genes are deterministic causes of ADAD, with virtually full penetrance, and a predictable age at symptomatic onset. Data and specimens collected are derived from full clinical assessments, including neurologic and physical examinations, extensive cognitive batteries, structural and functional neuro-imaging, amyloid and tau pathological measures using positron emission tomography (PET), flurordeoxyglucose (FDG) PET, cerebrospinal fluid and blood collection (plasma, serum, and whole blood), extensive genetic and multi-omic analyses, and brain donation upon death. This comprehensive evaluation of the human nervous system is performed longitudinally in both mutation carriers and family non-carriers, providing one of the deepest and broadest evaluations of the human brain across decades and through AD progression. These extensive data sets and samples are available for researchers to address scientific questions on the human brain, aging, and AD.
Collapse
Affiliation(s)
- Alisha J. Daniels
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Eric McDade
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | | | - Chengjie Xiong
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Richard J. Perrin
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Laura Ibanez
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | | | - Carlos Cruchaga
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Alison Goate
- Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Alan E. Renton
- Icahn School of Medicine at Mount Sinai, New York, NY USA
| | | | - Brian A. Gordon
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Jason Hassenstab
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Celeste Karch
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Brent Popp
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Allan Levey
- Goizueta Alzheimer’s Disease Research Center, Emory University, Atlanta, GA, USA
| | - John Morris
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Virginia Buckles
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | | | - Patricio Chrem
- Institute of Neurological Research FLENI, Buenos Aires, Argentina
| | | | - Jasmeer P. Chhatwal
- Massachusetts General and Brigham & Women’s Hospitals, Harvard Medical School, Boston MA, USA
| | | | - Nick C. Fox
- UK Dementia Research Institute at University College London, London, United Kingdom
- University College London, London, United Kingdom
| | | | - Takeshi Ikeuchi
- Brain Research Institute, Niigata University, Niigata, Japan
| | - Mathias Jucker
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- DZNE, German Center for Neurodegenerative Diseases, Tübingen, Germany
| | | | - Johannes Levin
- DZNE, German Center for Neurodegenerative Diseases, Munich, Germany
- Ludwig-Maximilians-Universität München, Munich, Germany
| | | | | | - Ana Luisa Sosa
- Instituto Nacional de Neurologia y Neurocirugla Innn, Mexico City, Mexico
| | - Ralph Martins
- Edith Cowan University, Western Australia, Australia
| | | | - James M. Noble
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Department of Neurology, and GH Sergievsky Center, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Edward Huey
- Brown University, Butler Hospital, Providence, RI, USA
| | - Pedro Rosa-Neto
- Centre de Recherche de L’hopital Douglas and McGill University, Montreal, Quebec
| | - Raquel Sánchez-Valle
- Hospital Clínic de Barcelona. IDIBAPS. University of Barcelona, Barcelona, Spain
| | - Peter R. Schofield
- Neuroscience Research Australia, Sydney, NSW, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Jee Hoon Roh
- Korea University, Korea University Anam Hospital, Seoul, South Korea
| | | | | |
Collapse
|
4
|
Dolatshahi M, Commean PK, Rahmani F, Liu J, Lloyd L, Nguyen C, Hantler N, Ly M, Yu G, Ippolito JE, Sirlin C, Morris JC, Benzinger TL, Raji CA. Alzheimer Disease Pathology and Neurodegeneration in Midlife Obesity: A Pilot Study. Aging Dis 2024; 15:1843-1854. [PMID: 37548931 PMCID: PMC11272197 DOI: 10.14336/ad.2023.0707] [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: 04/16/2023] [Accepted: 07/07/2023] [Indexed: 08/08/2023] Open
Abstract
Obesity and excess adiposity at midlife are risk factors for Alzheimer disease (AD). Visceral fat is known to be associated with insulin resistance and a pro-inflammatory state, the two mechanisms involved in AD pathology. We assessed the association of obesity, MRI-determined abdominal adipose tissue volumes, and insulin resistance with PET-determined amyloid and tau uptake in default mode network areas, and MRI-determined brain volume and cortical thickness in AD cortical signature in the cognitively normal midlife population. Thirty-two middle-aged (age: 51.27±6.12 years, 15 males, body mass index (BMI): 32.28±6.39 kg/m2) cognitively normal participants, underwent bloodwork, brain and abdominal MRI, and amyloid and tau PET scan. Visceral and subcutaneous adipose tissue (VAT, SAT) were semi-automatically segmented using VOXel Analysis Suite (Voxa). FreeSurfer was used to automatically segment brain regions using a probabilistic atlas. PET scans were acquired using [11C]PiB and AV-1451 tracers and were analyzed using PET unified pipeline. The association of brain volumes, cortical thicknesses, and PiB and AV-1451 standardized uptake value ratios (SUVRs) with BMI, VAT/SAT ratio, and insulin resistance were assessed using Spearman's partial correlation. VAT/SAT ratio was associated significantly with PiB SUVRs in the right precuneus cortex (p=0.034) overall, controlling for sex. This association was significant only in males (p=0.044), not females (p=0.166). Higher VAT/SAT ratio and PiB SUVRs in the right precuneus cortex were associated with lower cortical thickness in AD-signature areas predominantly including bilateral temporal cortices, parahippocampal, medial orbitofrontal, and cingulate cortices, with age and sex as covariates. Also, higher BMI and insulin resistance were associated with lower cortical thickness in bilateral temporal poles. In midlife cognitively normal adults, we demonstrated higher amyloid pathology in the right precuneus cortex in individuals with a higher VAT/SAT ratio, a marker of visceral obesity, along with a lower cortical thickness in AD-signature areas associated with higher visceral obesity, insulin resistance, and amyloid pathology.
Collapse
Affiliation(s)
- Mahsa Dolatshahi
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
| | - Paul K Commean
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
| | - Farzaneh Rahmani
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
| | - Jingxia Liu
- Washington University School of Medicine, Division of Public Health Sciences, Department of Surgery, St. Louis, Missouri, USA.
| | - LaKisha Lloyd
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
| | - Caitlyn Nguyen
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
| | - Nancy Hantler
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
| | - Maria Ly
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
| | - Gary Yu
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
| | - Joseph E Ippolito
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA.
| | - Claude Sirlin
- Liver Imaging Group, Department of Radiology, University of California, San Diego, La Jolla, California, USA.
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA.
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, Missouri, USA.
| | - Tammie L.S Benzinger
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, Missouri, USA.
- Department of Neurosurgery, Washington University School of Medicine, St Louis, Missouri, USA.
| | - Cyrus A Raji
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA.
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, Missouri, USA.
| |
Collapse
|
5
|
McDade E, Liu H, Bui Q, Hassenstab J, Gordon B, Benzinger T, Shen Y, Timsina J, Wang L, Sung YJ, Karch C, Renton A, Daniels A, Morris J, Xiong C, Ibanez L, Perrin R, Llibre-Guerra JJ, Day G, Supnet-Bell C, Xu X, Berman S, Chhatwal J, Ikeuchi T, Kasuga K, Niimi Y, Huey E, Schofield P, Brooks W, Ryan N, Jucker M, Laske C, Levin J, Vöglein J, Roh JH, Lopera F, Bateman R, Cruchaga C. Ubiquitin-Proteasome System in the Different Stages of Dominantly Inherited Alzheimer's Disease. RESEARCH SQUARE 2024:rs.3.rs-4202125. [PMID: 39108475 PMCID: PMC11302696 DOI: 10.21203/rs.3.rs-4202125/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
This study explored the role of the ubiquitin-proteasome system (UPS) in dominantly inherited Alzheimer's disease (DIAD) by examining changes in cerebrospinal fluid (CSF) levels of UPS proteins along with disease progression, AD imaging biomarkers (PiB PET, tau PET), neurodegeneration imaging measures (MRI, FDG PET), and Clinical Dementia Rating® (CDR®). Using the SOMAscan assay, we detected subtle increases in specific ubiquitin enzymes associated with proteostasis in mutation carriers (MCs) up to two decades before the estimated symptom onset. This was followed by more pronounced elevations of UPS-activating enzymes, including E2 and E3 proteins, and ubiquitin-related modifiers. Our findings also demonstrated consistent correlations between UPS proteins and CSF biomarkers such as Aβ42/40 ratio, total tau, various phosphorylated tau species to total tau ratios (ptau181/T181, ptauT205/T205, ptauS202/S202, ptauT217/T217), and MTBR-tau243, alongside Neurofilament light chain (NfL) and the CDR®. Notably, a positive association was observed with imaging markers (PiB PET, tau PET) and a negative correlation with markers of neurodegeneration (FDG PET, MRI), highlighting a significant link between UPS dysregulation and neurodegenerative processes. The correlations suggest that the increase in multiple UPS proteins with rising tau levels and tau-tangle associated markers, indicating a potential role for the UPS in relation to misfolded tau/neurofibrillary tangles (NFTs) and symptom onset. These findings indicate that elevated CSF UPS proteins in DIAD MCs could serve as early indicators of disease progression and suggest a link between UPS dysregulation and amyloid plaque, tau tangles formation, implicating the UPS as a potential therapeutic target in AD pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Alan Renton
- Nash Family Department of Neuroscience and Ronald Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA: Departments of Neurology and Genetics and Ge
| | | | | | | | | | | | | | | | | | | | | | - Jasmeer Chhatwal
- Massachusetts General Hospital, Brigham and Women's Hospital, Harvard Medical School
| | | | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University
| | | | | | | | | | | | | | | | | | | | | | | | - Randall Bateman
- Department of Neurology, Washington University School of Medicine
| | | |
Collapse
|
6
|
Kumar S, Earnest T, Yang B, Kothapalli D, Aschenbrenner AJ, Hassenstab J, Xiong C, Ances B, Morris J, Benzinger TLS, Gordon BA, Payne P, Sotiras A. Analyzing heterogeneity in Alzheimer Disease using multimodal normative modeling on imaging-based ATN biomarkers. ARXIV 2024:arXiv:2404.05748v2. [PMID: 39010871 PMCID: PMC11247918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
INTRODUCTION Previous studies have applied normative modeling on a single neuroimaging modality to investigate Alzheimer Disease (AD) heterogeneity. We employed a deep learning-based multimodal normative framework to analyze individual-level variation across ATN (amyloid-tau-neurodegeneration) imaging biomarkers. METHODS We selected cross-sectional discovery (n = 665) and replication cohorts (n = 430) with available T1-weighted MRI, amyloid and tau PET. Normative modeling estimated individual-level abnormal deviations in amyloid-positive individuals compared to amyloid-negative controls. Regional abnormality patterns were mapped at different clinical group levels to assess intra-group heterogeneity. An individual-level disease severity index (DSI) was calculated using both the spatial extent and magnitude of abnormal deviations across ATN. RESULTS Greater intra-group heterogeneity in ATN abnormality patterns was observed in more severe clinical stages of AD. Higher DSI was associated with worse cognitive function and increased risk of disease progression. DISCUSSION Subject-specific abnormality maps across ATN reveal the heterogeneous impact of AD on the brain.
Collapse
Affiliation(s)
- Sayantan Kumar
- Department of Computer Science and Engineering, Washington University in St Louis; 1 Brookings Drive, Saint Louis, MO 63130
- Institute for Informatics, Data Science & Biostatistics, Washington University School of Medicine in St Louis; 660 S. Euclid Ave, Campus Box 8132, Saint Louis, MO 63110
| | - Tom Earnest
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis; 4525 Scott Ave, Saint Louis, MO 63110
| | - Braden Yang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis; 4525 Scott Ave, Saint Louis, MO 63110
| | - Deydeep Kothapalli
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis; 4525 Scott Ave, Saint Louis, MO 63110
| | - Andrew J. Aschenbrenner
- Department of Neurology, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8111, St louis, MO 63110
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8111, St louis, MO 63110
| | - Chengie Xiong
- Institute for Informatics, Data Science & Biostatistics, Washington University School of Medicine in St Louis; 660 S. Euclid Ave, Campus Box 8132, Saint Louis, MO 63110
| | - Beau Ances
- Department of Neurology, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8111, St louis, MO 63110
| | - John Morris
- Department of Neurology, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8111, St louis, MO 63110
| | - Tammie L. S. Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis; 4525 Scott Ave, Saint Louis, MO 63110
| | - Brian A. Gordon
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis; 4525 Scott Ave, Saint Louis, MO 63110
| | - Philip Payne
- Department of Computer Science and Engineering, Washington University in St Louis; 1 Brookings Drive, Saint Louis, MO 63130
- Institute for Informatics, Data Science & Biostatistics, Washington University School of Medicine in St Louis; 660 S. Euclid Ave, Campus Box 8132, Saint Louis, MO 63110
| | - Aristeidis Sotiras
- Institute for Informatics, Data Science & Biostatistics, Washington University School of Medicine in St Louis; 660 S. Euclid Ave, Campus Box 8132, Saint Louis, MO 63110
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis; 4525 Scott Ave, Saint Louis, MO 63110
| |
Collapse
|
7
|
McKay NS, Millar PR, Nicosia J, Aschenbrenner AJ, Gordon BA, Benzinger TLS, Cruchaga CC, Schindler SE, Morris JC, Hassenstab J. Pick a PACC: Comparing domain-specific and general cognitive composites in Alzheimer disease research. Neuropsychology 2024; 38:443-464. [PMID: 38602816 PMCID: PMC11176005 DOI: 10.1037/neu0000949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024] Open
Abstract
OBJECTIVE We aimed to illustrate how complex cognitive data can be used to create domain-specific and general cognitive composites relevant to Alzheimer disease research. METHOD Using equipercentile equating, we combined data from the Charles F. and Joanne Knight Alzheimer Disease Research Center that spanned multiple iterations of the Uniform Data Set. Exploratory factor analyses revealed four domain-specific composites representing episodic memory, semantic memory, working memory, and attention/processing speed. The previously defined preclinical Alzheimer disease cognitive composite (PACC) and a novel alternative, the Knight-PACC, were also computed alongside a global composite comprising all available tests. These three composites allowed us to compare the usefulness of domain and general composites in the context of predicting common Alzheimer disease biomarkers. RESULTS General composites slightly outperformed domain-specific metrics in predicting imaging-derived amyloid, tau, and neurodegeneration burden. Power analyses revealed that the global, Knight-PACC, and attention and processing speed composites would require the smallest sample sizes to detect cognitive change in a clinical trial, while the Alzheimer Disease Cooperative Study-PACC required two to three times as many participants. CONCLUSIONS Analyses of cognition with the Knight-PACC and our domain-specific composites offer researchers flexibility by providing validated outcome assessments that can equate across test versions to answer a wide range of questions regarding cognitive decline in normal aging and neurodegenerative disease. (PsycInfo Database Record (c) 2024 APA, all rights reserved).
Collapse
|
8
|
Babulal GM, Chen L, Murphy SA, Carr DB, Morris JC. Predicting Driving Cessation Among Cognitively Normal Older Drivers: The Role of Alzheimer Disease Biomarkers and Clinical Assessments. Neurology 2024; 102:e209426. [PMID: 38776513 PMCID: PMC11226325 DOI: 10.1212/wnl.0000000000209426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/11/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND AND OBJECTIVES With the aging US population and increasing incidence of Alzheimer disease (AD), understanding factors contributing to driving cessation among older adults is crucial for clinicians. Driving is integral for maintaining independence and functional mobility, but the risk factors for driving cessation, particularly in the context of normal aging and preclinical AD, are not well understood. We studied a well-characterized community cohort to examine factors associated with driving cessation. METHODS This prospective, longitudinal observation study enrolled participants from the Knight Alzheimer Disease Research Center and The DRIVES Project. Participants were enrolled if they were aged 65 years or older, drove weekly, and were cognitively normal (Clinical Dementia Rating [CDR] = 0) at baseline. Participants underwent annual clinical, neurologic, and neuropsychological assessments, including β-amyloid PET imaging and CSF (Aβ42, total tau [t-Tau], and phosphorylated tau [p-Tau]) collection every 2-3 years. The primary outcome was time from baseline visit to driving cessation, accounting for death as a competing risk. The cumulative incidence function of driving cessation was estimated for each biomarker. The Fine and Gray subdistribution hazard model was used to examine the association between time to driving cessation and biomarkers adjusting for clinical and demographic covariates. RESULTS Among the 283 participants included in this study, there was a mean follow-up of 5.62 years. Driving cessation (8%) was associated with older age, female sex, progression to symptomatic AD (CDR ≥0.5), and poorer performance on a preclinical Alzheimer cognitive composite (PACC) score. Aβ PET imaging did not independently predict driving cessation, whereas CSF biomarkers, specifically t-Tau/Aβ42 (hazard ratio [HR] 2.82, 95% CI 1.23-6.44, p = 0.014) and p-Tau/Aβ42 (HR 2.91, 95% CI 1.28-6.59, p = 0.012) ratios, were independent predictors in the simple model adjusting for age, education, and sex. However, in the full model, progression to cognitive impairment based on the CDR and PACC score across each model was associated with a higher risk of driving cessation, whereas AD biomarkers were not statistically significant. DISCUSSION Female sex, CDR progression, and neuropsychological measures of cognitive functioning obtained in the clinic were strongly associated with future driving cessation. The results emphasize the need for early planning and conversations about driving retirement in the context of cognitive decline and the immense value of clinical measures in determining functional outcomes.
Collapse
Affiliation(s)
- Ganesh M Babulal
- From the Department of Neurology (G.M.B., S.A.M., J.C.M.), Division of Biostatistics (L.C.), and Department of Medicine (D.B.C.), Washington University School of Medicine, St. Louis, MO
| | - Ling Chen
- From the Department of Neurology (G.M.B., S.A.M., J.C.M.), Division of Biostatistics (L.C.), and Department of Medicine (D.B.C.), Washington University School of Medicine, St. Louis, MO
| | - Samantha A Murphy
- From the Department of Neurology (G.M.B., S.A.M., J.C.M.), Division of Biostatistics (L.C.), and Department of Medicine (D.B.C.), Washington University School of Medicine, St. Louis, MO
| | - David B Carr
- From the Department of Neurology (G.M.B., S.A.M., J.C.M.), Division of Biostatistics (L.C.), and Department of Medicine (D.B.C.), Washington University School of Medicine, St. Louis, MO
| | - John C Morris
- From the Department of Neurology (G.M.B., S.A.M., J.C.M.), Division of Biostatistics (L.C.), and Department of Medicine (D.B.C.), Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
9
|
O'Donnell JL, Soda AK, Jiang H, Norris SA, Maiti B, Karimi M, Campbell MC, Moerlein SM, Tu Z, Perlmutter JS. PET Quantification of [ 18F]VAT in Human Brain and Its Test-Retest Reproducibility and Age Dependence. J Nucl Med 2024; 65:956-961. [PMID: 38604762 PMCID: PMC11149597 DOI: 10.2967/jnumed.123.266860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 03/13/2024] [Indexed: 04/13/2024] Open
Abstract
Molecular imaging of brain vesicular acetylcholine transporter provides a biomarker to explore cholinergic systems in humans. We aimed to characterize the distribution of, and optimize methods to quantify, the vesicular acetylcholine transporter-specific tracer (-)-(1-(8-(2-[18F]fluoroethoxy)-3-hydroxy-1,2,3,4-tetrahydronaphthalen-2-yl)-piperidin-4-yl)(4-fluorophenyl)methanone ([18F]VAT) in the brain using PET. Methods: Fifty-two healthy participants aged 21-97 y had brain PET with [18F]VAT. [3H]VAT autoradiography identified brain areas devoid of specific binding in cortical white matter. PET image-based white matter reference region size, model start time, and duration were optimized for calculations of Logan nondisplaceable binding potential (BPND). Ten participants had 2 scans to determine test-retest variability. Finally, we analyzed age-dependent differences in participants. Results: [18F]VAT was widely distributed in the brain, with high striatal, thalamic, amygdala, hippocampal, cerebellar vermis, and regionally specific uptake in the cerebral cortex. [3H]VAT autoradiography-specific binding and PET [18F]VAT uptake were low in white matter. [18F]VAT SUVs in the white matter reference region correlated with age, requiring stringent erosion parameters. Logan BPND estimates stabilized using at least 40 min of data starting 25 min after injection. Test-retest variability had excellent reproducibility and reliability in repeat BPND calculations for 10 participants (putamen, 6.8%; r > 0.93). We observed age-dependent decreases in the caudate and putamen (multiple comparisons corrected) and in numerous cortical regions. Finally, we provide power tables to indicate potential mean differences that can be detected between 2 groups of participants. Conclusion: These results validate a reference region for BPND calculations and demonstrate the viability, reproducibility, and utility of using the [18F]VAT tracer in humans to quantify cholinergic pathways.
Collapse
Affiliation(s)
- John L O'Donnell
- Neurology, Washington University in Saint Louis, St. Louis, Missouri;
| | - Anil Kumar Soda
- Radiology, Washington University in Saint Louis, St. Louis, Missouri
| | - Hao Jiang
- Radiology, Washington University in Saint Louis, St. Louis, Missouri
| | - Scott A Norris
- Neurology, Washington University in Saint Louis, St. Louis, Missouri
- Radiology, Washington University in Saint Louis, St. Louis, Missouri
| | - Baijayanta Maiti
- Neurology, Washington University in Saint Louis, St. Louis, Missouri
- Radiology, Washington University in Saint Louis, St. Louis, Missouri
| | - Morvarid Karimi
- Neurology, Washington University in Saint Louis, St. Louis, Missouri
- Radiology, Washington University in Saint Louis, St. Louis, Missouri
| | - Meghan C Campbell
- Neurology, Washington University in Saint Louis, St. Louis, Missouri
- Radiology, Washington University in Saint Louis, St. Louis, Missouri
| | - Stephen M Moerlein
- Radiology, Washington University in Saint Louis, St. Louis, Missouri
- Biochemistry and Molecular Biophysics, Washington University in Saint Louis, St. Louis, Missouri; and
| | - Zhude Tu
- Radiology, Washington University in Saint Louis, St. Louis, Missouri
| | - Joel S Perlmutter
- Neurology, Washington University in Saint Louis, St. Louis, Missouri
- Radiology, Washington University in Saint Louis, St. Louis, Missouri
- Neuroscience, Physical, and Occupational Therapy, Washington University in Saint Louis, St. Louis, Missouri
| |
Collapse
|
10
|
Earnest T, Bani A, Ha SM, Hobbs DA, Kothapalli D, Yang B, Lee JJ, Benzinger TLS, Gordon BA, Sotiras A. Data-driven decomposition and staging of flortaucipir uptake in Alzheimer's disease. Alzheimers Dement 2024; 20:4002-4019. [PMID: 38683905 PMCID: PMC11180875 DOI: 10.1002/alz.13769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION Previous approaches pursuing in vivo staging of tau pathology in Alzheimer's disease (AD) have typically relied on neuropathologically defined criteria. In using predefined systems, these studies may miss spatial deposition patterns which are informative of disease progression. METHODS We selected discovery (n = 418) and replication (n = 132) cohorts with flortaucipir imaging. Non-negative matrix factorization (NMF) was applied to learn tau covariance patterns and develop a tau staging system. Flortaucipir components were also validated by comparison with amyloid burden, gray matter loss, and the expression of AD-related genes. RESULTS We found eight flortaucipir covariance patterns which were reproducible and overlapped with relevant gene expression maps. Tau stages were associated with AD severity as indexed by dementia status and neuropsychological performance. Comparisons of flortaucipir uptake with amyloid and atrophy also supported our model of tau progression. DISCUSSION Data-driven decomposition of flortaucipir uptake provides a novel framework for tau staging which complements existing systems. HIGHLIGHTS NMF reveals patterns of tau deposition in AD. Data-driven staging of flortaucipir tracks AD severity. Learned flortaucipir patterns overlap with AD-related gene expression.
Collapse
Affiliation(s)
- Tom Earnest
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Abdalla Bani
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Sung Min Ha
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Diana A. Hobbs
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Deydeep Kothapalli
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Braden Yang
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - John J. Lee
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Tammie L. S. Benzinger
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Brian A. Gordon
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Aristeidis Sotiras
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
- Institute for Informatics, Data Science & BiostatisticsWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | | |
Collapse
|
11
|
Li Y, Yen D, Hendrix RD, Gordon BA, Dlamini S, Barthélemy NR, Aschenbrenner AJ, Henson RL, Herries EM, Volluz K, Kirmess K, Eastwood S, Meyer M, Heller M, Jarrett L, McDade E, Holtzman DM, Benzinger TL, Morris JC, Bateman RJ, Xiong C, Schindler SE. Timing of Biomarker Changes in Sporadic Alzheimer's Disease in Estimated Years from Symptom Onset. Ann Neurol 2024; 95:951-965. [PMID: 38400792 PMCID: PMC11060905 DOI: 10.1002/ana.26891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/26/2023] [Accepted: 01/30/2024] [Indexed: 02/26/2024]
Abstract
OBJECTIVE A clock relating amyloid positron emission tomography (PET) to time was used to estimate the timing of biomarker changes in sporadic Alzheimer disease (AD). METHODS Research participants were included who underwent cerebrospinal fluid (CSF) collection within 2 years of amyloid PET. The ages at amyloid onset and AD symptom onset were estimated for each individual. The timing of change for plasma, CSF, imaging, and cognitive measures was calculated by comparing restricted cubic splines of cross-sectional data from the amyloid PET positive and negative groups. RESULTS The amyloid PET positive sub-cohort (n = 118) had an average age of 70.4 ± 7.4 years (mean ± standard deviation) and 16% were cognitively impaired. The amyloid PET negative sub-cohort (n = 277) included individuals with low levels of amyloid plaque burden at all scans who were cognitively unimpaired at the time of the scans. Biomarker changes were detected 15-19 years before estimated symptom onset for CSF Aβ42/Aβ40, plasma Aβ42/Aβ40, CSF pT217/T217, and amyloid PET; 12-14 years before estimated symptom onset for plasma pT217/T217, CSF neurogranin, CSF SNAP-25, CSF sTREM2, plasma GFAP, and plasma NfL; and 7-9 years before estimated symptom onset for CSF pT205/T205, CSF YKL-40, hippocampal volumes, and cognitive measures. INTERPRETATION The use of an amyloid clock enabled visualization and analysis of biomarker changes as a function of estimated years from symptom onset in sporadic AD. This study demonstrates that estimated years from symptom onset based on an amyloid clock can be used as a continuous staging measure for sporadic AD and aligns with findings in autosomal dominant AD. ANN NEUROL 2024;95:951-965.
Collapse
Affiliation(s)
- Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel Yen
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel D. Hendrix
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A. Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sibonginkhosi Dlamini
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicolas R. Barthélemy
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Rachel L. Henson
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Elizabeth M. Herries
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Katherine Volluz
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | - Maren Heller
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lea Jarrett
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Eric McDade
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - David M. Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L.S. Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J. Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Chengjie Xiong
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Suzanne E. Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
12
|
Doering S, McCullough A, Gordon BA, Chen CD, McKay N, Hobbs D, Keefe S, Flores S, Scott J, Smith H, Jarman S, Jackson K, Hornbeck RC, Ances BM, Xiong C, Aschenbrenner AJ, Hassenstab J, Cruchaga C, Daniels A, Bateman RJ, Morris JC, Benzinger TLS. Deconstructing pathological tau by biological process in early stages of Alzheimer disease: a method for quantifying tau spatial spread in neuroimaging. EBioMedicine 2024; 103:105080. [PMID: 38552342 PMCID: PMC10995809 DOI: 10.1016/j.ebiom.2024.105080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Neuroimaging studies often quantify tau burden in standardized brain regions to assess Alzheimer disease (AD) progression. However, this method ignores another key biological process in which tau spreads to additional brain regions. We have developed a metric for calculating the extent tau pathology has spread throughout the brain and evaluate the relationship between this metric and tau burden across early stages of AD. METHODS 445 cross-sectional participants (aged ≥ 50) who had MRI, amyloid PET, tau PET, and clinical testing were separated into disease-stage groups based on amyloid positivity and cognitive status (older cognitively normal control, preclinical AD, and symptomatic AD). Tau burden and tau spatial spread were calculated for all participants. FINDINGS We found both tau metrics significantly elevated across increasing disease stages (p < 0.0001) and as a function of increasing amyloid burden for participants with preclinical (p < 0.0001, p = 0.0056) and symptomatic (p = 0.010, p = 0.0021) AD. An interaction was found between tau burden and tau spatial spread when predicting amyloid burden (p = 0.00013). Analyses of slope between tau metrics demonstrated more spread than burden in preclinical AD (β = 0.59), but then tau burden elevated relative to spread (β = 0.42) once participants had symptomatic AD, when the tau metrics became highly correlated (R = 0.83). INTERPRETATION Tau burden and tau spatial spread are both strong biomarkers for early AD but provide unique information, particularly at the preclinical stage. Tau spatial spread may demonstrate earlier changes than tau burden which could have broad impact in clinical trial design. FUNDING This research was supported by the Knight Alzheimer Disease Research Center (Knight ADRC, NIH grants P30AG066444, P01AG026276, P01AG003991), Dominantly Inherited Alzheimer Network (DIAN, NIH grants U01AG042791, U19AG03243808, R01AG052550-01A1, R01AG05255003), and the Barnes-Jewish Hospital Foundation Willman Scholar Fund.
Collapse
Affiliation(s)
- Stephanie Doering
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Austin McCullough
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Brian A Gordon
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Charles D Chen
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Nicole McKay
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Diana Hobbs
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Sarah Keefe
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Shaney Flores
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Jalen Scott
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Hunter Smith
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Stephen Jarman
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Kelley Jackson
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Russ C Hornbeck
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Beau M Ances
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Chengjie Xiong
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | | | - Jason Hassenstab
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Carlos Cruchaga
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Alisha Daniels
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Randall J Bateman
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - John C Morris
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | | |
Collapse
|
13
|
Wisch JK, McKay NS, Boerwinkle AH, Kennedy J, Flores S, Handen BL, Christian BT, Head E, Mapstone M, Rafii MS, O'Bryant SE, Price JC, Laymon CM, Krinsky-McHale SJ, Lai F, Rosas HD, Hartley SL, Zaman S, Lott IT, Tudorascu D, Zammit M, Brickman AM, Lee JH, Bird TD, Cohen A, Chrem P, Daniels A, Chhatwal JP, Cruchaga C, Ibanez L, Jucker M, Karch CM, Day GS, Lee JH, Levin J, Llibre-Guerra J, Li Y, Lopera F, Roh JH, Ringman JM, Supnet-Bell C, van Dyck CH, Xiong C, Wang G, Morris JC, McDade E, Bateman RJ, Benzinger TLS, Gordon BA, Ances BM. Comparison of tau spread in people with Down syndrome versus autosomal-dominant Alzheimer's disease: a cross-sectional study. Lancet Neurol 2024; 23:500-510. [PMID: 38631766 PMCID: PMC11209765 DOI: 10.1016/s1474-4422(24)00084-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/01/2024] [Accepted: 02/21/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND In people with genetic forms of Alzheimer's disease, such as in Down syndrome and autosomal-dominant Alzheimer's disease, pathological changes specific to Alzheimer's disease (ie, accumulation of amyloid and tau) occur in the brain at a young age, when comorbidities related to ageing are not present. Studies including these cohorts could, therefore, improve our understanding of the early pathogenesis of Alzheimer's disease and be useful when designing preventive interventions targeted at disease pathology or when planning clinical trials. We compared the magnitude, spatial extent, and temporal ordering of tau spread in people with Down syndrome and autosomal-dominant Alzheimer's disease. METHODS In this cross-sectional observational study, we included participants (aged ≥25 years) from two cohort studies. First, we collected data from the Dominantly Inherited Alzheimer's Network studies (DIAN-OBS and DIAN-TU), which include carriers of autosomal-dominant Alzheimer's disease genetic mutations and non-carrier familial controls recruited in Australia, Europe, and the USA between 2008 and 2022. Second, we collected data from the Alzheimer Biomarkers Consortium-Down Syndrome study, which includes people with Down syndrome and sibling controls recruited from the UK and USA between 2015 and 2021. Controls from the two studies were combined into a single group of familial controls. All participants had completed structural MRI and tau PET (18F-flortaucipir) imaging. We applied Gaussian mixture modelling to identify regions of high tau PET burden and regions with the earliest changes in tau binding for each cohort separately. We estimated regional tau PET burden as a function of cortical amyloid burden for both cohorts. Finally, we compared the temporal pattern of tau PET burden relative to that of amyloid. FINDINGS We included 137 people with Down syndrome (mean age 38·5 years [SD 8·2], 74 [54%] male, and 63 [46%] female), 49 individuals with autosomal-dominant Alzheimer's disease (mean age 43·9 years [11·2], 22 [45%] male, and 27 [55%] female), and 85 familial controls, pooled from across both studies (mean age 41·5 years [12·1], 28 [33%] male, and 57 [67%] female), who satisfied the PET quality-control procedure for tau-PET imaging processing. 134 (98%) people with Down syndrome, 44 (90%) with autosomal-dominant Alzheimer's disease, and 77 (91%) controls also completed an amyloid PET scan within 3 years of tau PET imaging. Spatially, tau PET burden was observed most frequently in subcortical and medial temporal regions in people with Down syndrome, and within the medial temporal lobe in people with autosomal-dominant Alzheimer's disease. Across the brain, people with Down syndrome had greater concentrations of tau for a given level of amyloid compared with people with autosomal-dominant Alzheimer's disease. Temporally, increases in tau were more strongly associated with increases in amyloid for people with Down syndrome compared with autosomal-dominant Alzheimer's disease. INTERPRETATION Although the general progression of amyloid followed by tau is similar for people Down syndrome and people with autosomal-dominant Alzheimer's disease, we found subtle differences in the spatial distribution, timing, and magnitude of the tau burden between these two cohorts. These differences might have important implications; differences in the temporal pattern of tau accumulation might influence the timing of drug administration in clinical trials, whereas differences in the spatial pattern and magnitude of tau burden might affect disease progression. FUNDING None.
Collapse
Affiliation(s)
- Julie K Wisch
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA.
| | - Nicole S McKay
- Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Anna H Boerwinkle
- McGovern Medical School, University of Texas in Houston, Houston, TX, USA
| | - James Kennedy
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Shaney Flores
- Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Benjamin L Handen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bradley T Christian
- Department of Medical Physics and Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth Head
- Department of Pathology, Gillespie Neuroscience Research Facility, University of California, Irvine, CA, USA
| | - Mark Mapstone
- Department of Neurology, University of California Irvine School of Medicine, Irvine, CA, USA
| | - Michael S Rafii
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Sid E O'Bryant
- Institute for Translational Research Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Julie C Price
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Charles M Laymon
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sharon J Krinsky-McHale
- Department of Psychology, New York State Institute for Basic Research in Developmental Disabilities, New York, NY, USA
| | - Florence Lai
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - H Diana Rosas
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA; Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Sigan L Hartley
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Shahid Zaman
- Cambridge Intellectual and Developmental Disabilities Research Group, University of Cambridge, Cambridge, UK
| | - Ira T Lott
- Department of Pediatrics, University of California Irvine School of Medicine, Irvine, CA, USA
| | - Dana Tudorascu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew Zammit
- Department of Medical Physics and Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| | - Adam M Brickman
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Joseph H Lee
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA; Department of Epidemiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Thomas D Bird
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Annie Cohen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patricio Chrem
- Centro de Memoria y Envejecimiento, Buenos Aires, Argentina
| | - Alisha Daniels
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA; Hope Center for Neurological Disorders, Washington University in St Louis, St Louis, MO, USA
| | - Laura Ibanez
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Mathias Jucker
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Celeste M Karch
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA; Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA; German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Gregory S Day
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Jae-Hong Lee
- Department of Neurology, University of Ulsan College of Medicine, Asian Medical Center, Seoul, South Korea
| | - Johannes Levin
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases, site Munich, Munich, Germany; Munich Cluster for Systems Neurology, Munich, Germany
| | - Jorge Llibre-Guerra
- Hope Center for Neurological Disorders, Washington University in St Louis, St Louis, MO, USA
| | - Yan Li
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA; Department of Biostatistics, Washington University in St Louis, St Louis, MO, USA
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Jee Hoon Roh
- Departments of Physiology and Neurology, Korea University College of Medicine, Seoul, South Korea
| | - John M Ringman
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of USC, Los Angeles, CA, USA
| | | | | | - Chengjie Xiong
- Department of Biostatistics, Washington University in St Louis, St Louis, MO, USA
| | - Guoqiao Wang
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA; Department of Biostatistics, Washington University in St Louis, St Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Eric McDade
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Randall J Bateman
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | | | - Brian A Gordon
- Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Beau M Ances
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| |
Collapse
|
14
|
Wang G, Li Y, Xiong C, Benzinger TLS, Gordon BA, Hassenstab J, Aschenbrenner AJ, McDade E, Clifford DB, Libre‐Guerra JJ, Shi X, Mummery CJ, van Dyck CH, Lah JJ, Honig LS, Day G, Ringman JM, Brooks WS, Fox NC, Suzuki K, Levin J, Jucker M, Delmar P, Bittner T, Bateman RJ. Examining amyloid reduction as a surrogate endpoint through latent class analysis using clinical trial data for dominantly inherited Alzheimer's disease. Alzheimers Dement 2024; 20:2698-2706. [PMID: 38400532 PMCID: PMC11032558 DOI: 10.1002/alz.13735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/18/2023] [Accepted: 01/18/2024] [Indexed: 02/25/2024]
Abstract
INTRODUCTION Increasing evidence suggests that amyloid reduction could serve as a plausible surrogate endpoint for clinical and cognitive efficacy. The double-blind phase 3 DIAN-TU-001 trial tested clinical and cognitive declines with increasing doses of solanezumab or gantenerumab. METHODS We used latent class (LC) analysis on data from the Dominantly Inherited Alzheimer Network Trials Unit 001 trial to test amyloid positron emission tomography (PET) reduction as a potential surrogate biomarker. RESULTS LC analysis categorized participants into three classes: amyloid no change, amyloid reduction, and amyloid growth, based on longitudinal amyloid Pittsburgh compound B PET standardized uptake value ratio data. The amyloid-no-change class was at an earlier disease stage for amyloid amounts and dementia. Despite similar baseline characteristics, the amyloid-reduction class exhibited reductions in the annual decline rates compared to the amyloid-growth class across multiple biomarker, clinical, and cognitive outcomes. DISCUSSION LC analysis indicates that amyloid reduction is associated with improved clinical outcomes and supports its use as a surrogate biomarker in clinical trials. HIGHLIGHTS We used latent class (LC) analysis to test amyloid reduction as a surrogate biomarker. Despite similar baseline characteristics, the amyloid-reduction class exhibited remarkably better outcomes compared to the amyloid-growth class across multiple measures. LC analysis proves valuable in testing amyloid reduction as a surrogate biomarker in clinical trials lacking significant treatment effects.
Collapse
Affiliation(s)
- Guoqiao Wang
- Washington University, School of MedicineSt. LouisMissouriUSA
| | - Yan Li
- Washington University, School of MedicineSt. LouisMissouriUSA
| | - Chengjie Xiong
- Washington University, School of MedicineSt. LouisMissouriUSA
| | | | - Brian A. Gordon
- Washington University, School of MedicineSt. LouisMissouriUSA
| | | | | | - Eric McDade
- Washington University, School of MedicineSt. LouisMissouriUSA
| | | | | | - Xinyu Shi
- Washington University, School of MedicineSt. LouisMissouriUSA
| | | | | | - James J. Lah
- Emory University Medical CenterAtlantaGeorgiaUSA
| | | | - Gregg Day
- Mayo Clinic JacksonvilleJacksonvilleFloridaUSA
| | - John M. Ringman
- Department of NeurologyKeck School of Medicine of USCLos AngelesCaliforniaUSA
| | - William S. Brooks
- Neuroscience Research Australia, Randwick NSW Australia, and School of Clinical MedicineUniversity of New South WalesRandwickNew South WalesAustralia
| | - Nick C. Fox
- Dementia Research CentreUniversity College LondonLondonUK
| | | | - Johannes Levin
- Department of NeurologyLudwig‐Maximilians‐Universität MünchenMunichGermany
- German Center for Neurodegenerative DiseasesMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Mathias Jucker
- Department of Cellular NeurologyHertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
| | | | - Tobias Bittner
- F.Hoffmann‐LaRoche, Ltd.BaselSwitzerland
- Genentech, Inc., a member of the Roche GroupSouth San FranciscoCaliforniaUSA
| | | | | |
Collapse
|
15
|
Joseph‐Mathurin N, Feldman RL, Lu R, Shirzadi Z, Toomer C, Saint Clair JR, Ma Y, McKay NS, Strain JF, Kilgore C, Friedrichsen KA, Chen CD, Gordon BA, Chen G, Hornbeck RC, Massoumzadeh P, McCullough AA, Wang Q, Li Y, Wang G, Keefe SJ, Schultz SA, Cruchaga C, Preboske GM, Jack CR, Llibre‐Guerra JJ, Allegri RF, Ances BM, Berman SB, Brooks WS, Cash DM, Day GS, Fox NC, Fulham M, Ghetti B, Johnson KA, Jucker M, Klunk WE, la Fougère C, Levin J, Niimi Y, Oh H, Perrin RJ, Reischl G, Ringman JM, Saykin AJ, Schofield PR, Su Y, Supnet‐Bell C, Vöglein J, Yakushev I, Brickman AM, Morris JC, McDade E, Xiong C, Bateman RJ, Chhatwal JP, Benzinger TLS. Presenilin-1 mutation position influences amyloidosis, small vessel disease, and dementia with disease stage. Alzheimers Dement 2024; 20:2680-2697. [PMID: 38380882 PMCID: PMC11032566 DOI: 10.1002/alz.13729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 02/22/2024]
Abstract
INTRODUCTION Amyloidosis, including cerebral amyloid angiopathy, and markers of small vessel disease (SVD) vary across dominantly inherited Alzheimer's disease (DIAD) presenilin-1 (PSEN1) mutation carriers. We investigated how mutation position relative to codon 200 (pre-/postcodon 200) influences these pathologic features and dementia at different stages. METHODS Individuals from families with known PSEN1 mutations (n = 393) underwent neuroimaging and clinical assessments. We cross-sectionally evaluated regional Pittsburgh compound B-positron emission tomography uptake, magnetic resonance imaging markers of SVD (diffusion tensor imaging-based white matter injury, white matter hyperintensity volumes, and microhemorrhages), and cognition. RESULTS Postcodon 200 carriers had lower amyloid burden in all regions but worse markers of SVD and worse Clinical Dementia Rating® scores compared to precodon 200 carriers as a function of estimated years to symptom onset. Markers of SVD partially mediated the mutation position effects on clinical measures. DISCUSSION We demonstrated the genotypic variability behind spatiotemporal amyloidosis, SVD, and clinical presentation in DIAD, which may inform patient prognosis and clinical trials. HIGHLIGHTS Mutation position influences Aβ burden, SVD, and dementia. PSEN1 pre-200 group had stronger associations between Aβ burden and disease stage. PSEN1 post-200 group had stronger associations between SVD markers and disease stage. PSEN1 post-200 group had worse dementia score than pre-200 in late disease stage. Diffusion tensor imaging-based SVD markers mediated mutation position effects on dementia in the late stage.
Collapse
|
16
|
Meeker KL, Luckett PH, Barthélemy NR, Hobbs DA, Chen C, Bollinger J, Ovod V, Flores S, Keefe S, Henson RL, Herries EM, McDade E, Hassenstab JJ, Xiong C, Cruchaga C, Benzinger TLS, Holtzman DM, Schindler SE, Bateman RJ, Morris JC, Gordon BA, Ances BM. Comparison of cerebrospinal fluid, plasma and neuroimaging biomarker utility in Alzheimer's disease. Brain Commun 2024; 6:fcae081. [PMID: 38505230 PMCID: PMC10950051 DOI: 10.1093/braincomms/fcae081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/01/2024] [Accepted: 03/14/2024] [Indexed: 03/21/2024] Open
Abstract
Alzheimer's disease biomarkers are crucial to understanding disease pathophysiology, aiding accurate diagnosis and identifying target treatments. Although the number of biomarkers continues to grow, the relative utility and uniqueness of each is poorly understood as prior work has typically calculated serial pairwise relationships on only a handful of markers at a time. The present study assessed the cross-sectional relationships among 27 Alzheimer's disease biomarkers simultaneously and determined their ability to predict meaningful clinical outcomes using machine learning. Data were obtained from 527 community-dwelling volunteers enrolled in studies at the Charles F. and Joanne Knight Alzheimer Disease Research Center at Washington University in St Louis. We used hierarchical clustering to group 27 imaging, CSF and plasma measures of amyloid beta, tau [phosphorylated tau (p-tau), total tau t-tau)], neuronal injury and inflammation drawn from MRI, PET, mass-spectrometry assays and immunoassays. Neuropsychological and genetic measures were also included. Random forest-based feature selection identified the strongest predictors of amyloid PET positivity across the entire cohort. Models also predicted cognitive impairment across the entire cohort and in amyloid PET-positive individuals. Four clusters emerged reflecting: core Alzheimer's disease pathology (amyloid and tau), neurodegeneration, AT8 antibody-associated phosphorylated tau sites and neuronal dysfunction. In the entire cohort, CSF p-tau181/Aβ40lumi and Aβ42/Aβ40lumi and mass spectrometry measurements for CSF pT217/T217, pT111/T111, pT231/T231 were the strongest predictors of amyloid PET status. Given their ability to denote individuals on an Alzheimer's disease pathological trajectory, these same markers (CSF pT217/T217, pT111/T111, p-tau/Aβ40lumi and t-tau/Aβ40lumi) were largely the best predictors of worse cognition in the entire cohort. When restricting analyses to amyloid-positive individuals, the strongest predictors of impaired cognition were tau PET, CSF t-tau/Aβ40lumi, p-tau181/Aβ40lumi, CSF pT217/217 and pT205/T205. Non-specific CSF measures of neuronal dysfunction and inflammation were poor predictors of amyloid PET and cognitive status. The current work utilized machine learning to understand the interrelationship structure and utility of a large number of biomarkers. The results demonstrate that, although the number of biomarkers has rapidly expanded, many are interrelated and few strongly predict clinical outcomes. Examining the entire corpus of available biomarkers simultaneously provides a meaningful framework to understand Alzheimer's disease pathobiological change as well as insight into which biomarkers may be most useful in Alzheimer's disease clinical practice and trials.
Collapse
Affiliation(s)
- Karin L Meeker
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Patrick H Luckett
- Department of Neurosurgery, Washington University in St Louis, St Louis, MO 63110, USA
| | - Nicolas R Barthélemy
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Diana A Hobbs
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Charles Chen
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
| | - James Bollinger
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Vitaliy Ovod
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Shaney Flores
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Sarah Keefe
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Rachel L Henson
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Elizabeth M Herries
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Eric McDade
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Jason J Hassenstab
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Chengjie Xiong
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
- Division of Biostatistics, Washington University in St Louis, St Louis, MO 63110, USA
| | - Carlos Cruchaga
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Randall J Bateman
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - John C Morris
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Brian A Gordon
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Beau M Ances
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|
17
|
Azimi MS, Kamali-Asl A, Ay MR, Zeraatkar N, Hosseini MS, Sanaat A, Dadgar H, Arabi H. Deep learning-based partial volume correction in standard and low-dose positron emission tomography-computed tomography imaging. Quant Imaging Med Surg 2024; 14:2146-2164. [PMID: 38545051 PMCID: PMC10963814 DOI: 10.21037/qims-23-871] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 11/20/2023] [Indexed: 08/05/2024]
Abstract
BACKGROUND Positron emission tomography (PET) imaging encounters the obstacle of partial volume effects, arising from its limited intrinsic resolution, giving rise to (I) considerable bias, particularly for structures comparable in size to the point spread function (PSF) of the system; and (II) blurred image edges and blending of textures along the borders. We set out to build a deep learning-based framework for predicting partial volume corrected full-dose (FD + PVC) images from either standard or low-dose (LD) PET images without requiring any anatomical data in order to provide a joint solution for partial volume correction and de-noise LD PET images. METHODS We trained a modified encoder-decoder U-Net network with standard of care or LD PET images as the input and FD + PVC images by six different PVC methods as the target. These six PVC approaches include geometric transfer matrix (GTM), multi-target correction (MTC), region-based voxel-wise correction (RBV), iterative Yang (IY), reblurred Van-Cittert (RVC), and Richardson-Lucy (RL). The proposed models were evaluated using standard criteria, such as peak signal-to-noise ratio (PSNR), root mean squared error (RMSE), structural similarity index (SSIM), relative bias, and absolute relative bias. RESULTS Different levels of error were observed for these partial volume correction methods, which were relatively smaller for GTM with a SSIM of 0.63 for LD and 0.29 for FD, IY with an SSIM of 0.63 for LD and 0.67 for FD, RBV with an SSIM of 0.57 for LD and 0.65 for FD, and RVC with an SSIM of 0.89 for LD and 0.94 for FD PVC approaches. However, large quantitative errors were observed for multi-target MTC with an RMSE of 2.71 for LD and 2.45 for FD and RL with an RMSE of 5 for LD and 3.27 for FD PVC approaches. CONCLUSIONS We found that the proposed framework could effectively perform joint de-noising and partial volume correction for PET images with LD and FD input PET data (LD vs. FD). When no magnetic resonance imaging (MRI) images are available, the developed deep learning models could be used for partial volume correction on LD or standard PET-computed tomography (PET-CT) scans as an image quality enhancement technique.
Collapse
Affiliation(s)
- Mohammad-Saber Azimi
- Department of Medical Radiation Engineering, Shahid Beheshti University, Tehran, Iran
- Research Center for Molecular and Cellular Imaging (RCMCI), Advanced Medical Technologies and Equipment Institute (AMTEI), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Alireza Kamali-Asl
- Department of Medical Radiation Engineering, Shahid Beheshti University, Tehran, Iran
| | - Mohammad-Reza Ay
- Research Center for Molecular and Cellular Imaging (RCMCI), Advanced Medical Technologies and Equipment Institute (AMTEI), Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Department of Medical Physics and Biomedical Engineering, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Amirhossein Sanaat
- Division of Nuclear Medicine & Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | - Habibollah Dadgar
- Cancer Research Center, Razavi Hospital, Imam Reza International University, Mashhad, Iran
| | - Hossein Arabi
- Division of Nuclear Medicine & Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| |
Collapse
|
18
|
Su Y, Protas H, Luo J, Chen K, Alosco ML, Adler CH, Balcer LJ, Bernick C, Au R, Banks SJ, Barr WB, Coleman MJ, Dodick DW, Katz DI, Marek KL, McClean MD, McKee AC, Mez J, Daneshvar DH, Palmisano JN, Peskind ER, Turner RW, Wethe JV, Rabinovici G, Johnson K, Tripodis Y, Cummings JL, Shenton ME, Stern RA, Reiman EM. Flortaucipir tau PET findings from former professional and college American football players in the DIAGNOSE CTE research project. Alzheimers Dement 2024; 20:1827-1838. [PMID: 38134231 PMCID: PMC10984430 DOI: 10.1002/alz.13602] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 10/27/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023]
Abstract
INTRODUCTION Tau is a key pathology in chronic traumatic encephalopathy (CTE). Here, we report our findings in tau positron emission tomography (PET) measurements from the DIAGNOSE CTE Research Project. METHOD We compare flortaucipir PET measures from 104 former professional players (PRO), 58 former college football players (COL), and 56 same-age men without exposure to repetitive head impacts (RHI) or traumatic brain injury (unexposed [UE]); characterize their associations with RHI exposure; and compare players who did or did not meet diagnostic criteria for traumatic encephalopathy syndrome (TES). RESULTS Significantly elevated flortaucipir uptake was observed in former football players (PRO+COL) in prespecified regions (p < 0.05). Association between regional flortaucipir uptake and estimated cumulative head impact exposure was only observed in the superior frontal region in former players over 60 years old. Flortaucipir PET was not able to differentiate TES groups. DISCUSSION Additional studies are needed to further understand tau pathology in CTE and other individuals with a history of RHI.
Collapse
Affiliation(s)
- Yi Su
- Banner Alzheimer's Institute and Arizona Alzheimer's ConsortiumPhoenixArizonaUSA
| | - Hillary Protas
- Banner Alzheimer's Institute and Arizona Alzheimer's ConsortiumPhoenixArizonaUSA
| | - Ji Luo
- Banner Alzheimer's Institute and Arizona Alzheimer's ConsortiumPhoenixArizonaUSA
| | - Kewei Chen
- Banner Alzheimer's Institute and Arizona Alzheimer's ConsortiumPhoenixArizonaUSA
| | - Michael L. Alosco
- Department of NeurologyBoston University Alzheimer's Disease Research CenterBoston University CTE CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Charles H. Adler
- Department of NeurologyMayo Clinic College of Medicine, Mayo Clinic ArizonaScottsdaleArizonaUSA
| | - Laura J. Balcer
- Departments of NeurologyNYU Grossman School of MedicineNew YorkNew YorkUSA
- Department of Population Health and OphthalmologyNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Charles Bernick
- Cleveland Clinic Lou Ruvo Center for Brain HealthLas VegasNevadaUSA
- Department of NeurologyUniversity of WashingtonSeattleWashingtonUSA
| | - Rhoda Au
- Department of NeurologyBoston University Alzheimer's Disease Research CenterBoston University CTE CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Framingham Heart StudyFraminghamMassachusettsUSA
- Slone Epidemiology Center; Departments of Anatomy & Neurobiology, Neurology, and MedicineDepartment of EpidemiologyBoston University Chobanian & Avedisian School of Medicine; Boston University School of Public HealthBostonMassachusettsUSA
| | - Sarah J. Banks
- Departments of Neuroscience and PsychiatryUniversity of CaliforniaSan DiegoCaliforniaUSA
| | - William B. Barr
- Departments of NeurologyNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Michael J. Coleman
- Departments of Psychiatry and RadiologyPsychiatry Neuroimaging LaboratoryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - David W. Dodick
- Department of NeurologyMayo Clinic College of Medicine, Mayo Clinic ArizonaScottsdaleArizonaUSA
| | - Douglas I. Katz
- Department of NeurologyBoston University Alzheimer's Disease Research CenterBoston University CTE CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Encompass Health Braintree Rehabilitation HospitalBraintreeMassachusettsUSA
| | - Kenneth L. Marek
- Institute for Neurodegenerative Disorders, Invicro, LLCNew HavenConnecticutUSA
| | - Michael D. McClean
- Department of Environmental HealthBoston University School of Public HealthBostonMassachusettsUSA
| | - Ann C. McKee
- Department of NeurologyBoston University Alzheimer's Disease Research CenterBoston University CTE CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- VA Boston Healthcare SystemBostonMassachusettsUSA
| | - Jesse Mez
- Department of NeurologyBoston University Alzheimer's Disease Research CenterBoston University CTE CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Framingham Heart StudyFraminghamMassachusettsUSA
| | - Daniel H. Daneshvar
- Department of Physical Medicine & RehabilitationMassachusetts General Hospital, Spaulding Rehabilitation Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Joseph N. Palmisano
- Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Biostatistics and Epidemiology Data Analytics Center (BEDAC), Boston University School of Public HealthBostonMassachusettsUSA
| | - Elaine R. Peskind
- Department of Psychiatry and Behavioral SciencesVA Northwest Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System; University of Washington School of MedicineSeattleWashingtonUSA
| | - Robert W. Turner
- Department of Clinical Research & LeadershipThe George Washington University School of Medicine & Health SciencesWashingtonDistrict of ColumbiaUSA
| | - Jennifer V. Wethe
- Department of Psychiatry and PsychologyMayo Clinic School of Medicine, Mayo Clinic ArizonaScottsdaleArizonaUSA
| | - Gil Rabinovici
- Department of NeurologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Keith Johnson
- Gordon Center for Medical Imaging, Mass General Research Institute, Harvard Medical SchoolBostonMassachusettsUSA
| | - Yorghos Tripodis
- Department of NeurologyBoston University Alzheimer's Disease Research CenterBoston University CTE CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Jeffrey L. Cummings
- Department of Brain HealthChambers‐Grundy Center for Transformative NeuroscienceSchool of Integrated Health Sciences, University of Nevada Las VegasLas VegasNevadaUSA
| | - Martha E. Shenton
- Departments of Psychiatry and RadiologyPsychiatry Neuroimaging LaboratoryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Robert A. Stern
- Department of NeurologyBoston University Alzheimer's Disease Research CenterBoston University CTE CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Eric M. Reiman
- Banner Alzheimer's Institute and Arizona Alzheimer's ConsortiumPhoenixArizonaUSA
- University of Arizona, Arizona State University, Translational Genomics Research InstitutePhoenixArizonaUSA
| | | |
Collapse
|
19
|
Chen G, McKay NS, Gordon BA, Liu J, Joseph-Mathurin N, Schindler SE, Hassenstab J, Aschenbrenner AJ, Wang Q, Schultz SA, Su Y, LaMontagne PJ, Keefe SJ, Massoumzadeh P, Cruchaga C, Xiong C, Morris JC, Benzinger TLS. Predicting cognitive decline: Which is more useful, baseline amyloid levels or longitudinal change? Neuroimage Clin 2023; 41:103551. [PMID: 38150745 PMCID: PMC10788301 DOI: 10.1016/j.nicl.2023.103551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 12/10/2023] [Indexed: 12/29/2023]
Abstract
The use of biomarkers for the early detection of Alzheimer's disease (AD) is crucial for developing potential therapeutic treatments. Positron Emission Tomography (PET) is a well-established tool used to detect β-amyloid (Aβ) plaques in the brain. Previous studies have shown that cross-sectional biomarkers can predict cognitive decline (Schindler et al.,2021). However, it is still unclear whether longitudinal Aβ-PET may have additional value for predicting time to cognitive impairment in AD. The current study aims to evaluate the ability of baseline- versus longitudinal rate of change in-11C-Pittsburgh compound B (PiB) Aβ-PET to predict cognitive decline. A cohort of 153 participants who previously underwent PiB-PET scans and comprehensive clinical assessments were used in this study. Our analyses revealed that baseline Aβ is significantly associated with the rate of change in cognitive composite scores, with cognition declining more rapidly when baseline PiB Aβ levels were higher. In contrast, no signification association was identified between the rate of change in PiB-PET Aβ and cognitive decline. Additionally, the ability of the rate of change in the PiB-PET measures to predict cognitive decline was significantly influenced by APOE ε4 carrier status. These results suggest that a single PiB-PET scan is sufficient to predict cognitive decline and that longitudinal measures of Aβ accumulation do not improve the prediction of cognitive decline once someone is amyloid positive.
Collapse
Affiliation(s)
- Gengsheng Chen
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Nicole S McKay
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Jingxia Liu
- Department of Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Nelly Joseph-Mathurin
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Andrew J Aschenbrenner
- Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Qing Wang
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Stephanie A Schultz
- Department of Neurology Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yi Su
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Pamela J LaMontagne
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Sarah J Keefe
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Parinaz Massoumzadeh
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Chengjie Xiong
- Divison of Biostatistics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
20
|
Rahmani F, Brier MR, Gordon BA, McKay N, Flores S, Keefe S, Hornbeck R, Ances B, Joseph‐Mathurin N, Xiong C, Wang G, Raji CA, Libre‐Guerra JJ, Perrin RJ, McDade E, Daniels A, Karch C, Day GS, Brickman AM, Fulham M, Jack CR, la La Fougère C, Reischl G, Schofield PR, Oh H, Levin J, Vöglein J, Cash DM, Yakushev I, Ikeuchi T, Klunk WE, Morris JC, Bateman RJ, Benzinger TLS. T1 and FLAIR signal intensities are related to tau pathology in dominantly inherited Alzheimer disease. Hum Brain Mapp 2023; 44:6375-6387. [PMID: 37867465 PMCID: PMC10681640 DOI: 10.1002/hbm.26514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/17/2023] [Accepted: 09/27/2023] [Indexed: 10/24/2023] Open
Abstract
Carriers of mutations responsible for dominantly inherited Alzheimer disease provide a unique opportunity to study potential imaging biomarkers. Biomarkers based on routinely acquired clinical MR images, could supplement the extant invasive or logistically challenging) biomarker studies. We used 1104 longitudinal MR, 324 amyloid beta, and 87 tau positron emission tomography imaging sessions from 525 participants enrolled in the Dominantly Inherited Alzheimer Network Observational Study to extract novel imaging metrics representing the mean (μ) and standard deviation (σ) of standardized image intensities of T1-weighted and Fluid attenuated inversion recovery (FLAIR) MR scans. There was an exponential decrease in FLAIR-μ in mutation carriers and an increase in FLAIR and T1 signal heterogeneity (T1-σ and FLAIR-σ) as participants approached the symptom onset in both supramarginal, the right postcentral and right superior temporal gyri as well as both caudate nuclei, putamina, thalami, and amygdalae. After controlling for the effect of regional atrophy, FLAIR-μ decreased and T1-σ and FLAIR-σ increased with increasing amyloid beta and tau deposition in numerous cortical regions. In symptomatic mutation carriers and independent of the effect of regional atrophy, tau pathology demonstrated a stronger relationship with image intensity metrics, compared with amyloid pathology. We propose novel MR imaging intensity-based metrics using standard clinical T1 and FLAIR images which strongly associates with the progression of pathology in dominantly inherited Alzheimer disease. We suggest that tau pathology may be a key driver of the observed changes in this cohort of patients.
Collapse
Affiliation(s)
| | | | - Brian A. Gordon
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Nicole McKay
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Shaney Flores
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Sarah Keefe
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Russ Hornbeck
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Beau Ances
- Washington University School of MedicineSt. LouisMissouriUSA
| | | | - Chengjie Xiong
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Guoqiao Wang
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Cyrus A. Raji
- Washington University School of MedicineSt. LouisMissouriUSA
| | | | | | - Eric McDade
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Alisha Daniels
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Celeste Karch
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Gregory S. Day
- Mayo Clinic, Department of NeurologyJacksonvilleFloridaUSA
| | - Adam M. Brickman
- Taub Institute for Research on Alzheimer's Disease & the Aging Brain, and Department of Neurology College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | | | | | - Christian la La Fougère
- Department of Nuclear Medicine and Clinical Molecular ImagingUniversity Hospital TuebingenTübingenGermany
- German Center for Neurodegenerative Diseases (DZNE) TuebingenTübingenGermany
- Department of Preclinical Imaging and RadiopharmacyEberhard Karls University TübingenTübingenGermany
| | - Gerald Reischl
- Department of Nuclear Medicine and Clinical Molecular ImagingUniversity Hospital TuebingenTübingenGermany
- German Center for Neurodegenerative Diseases (DZNE) TuebingenTübingenGermany
- Department of Preclinical Imaging and RadiopharmacyEberhard Karls University TübingenTübingenGermany
| | - Peter R. Schofield
- Neuroscience Research AustraliaSydneyNew South WalesAustralia
- School of Biomedical SciencesUniversity of New South WalesSydneyNew South WalesAustralia
| | - Hwamee Oh
- Brown UniversityProvidenceRhode IslandUSA
| | - Johannes Levin
- Department of NeurologyLudwig‐Maximilians‐Universität MünchenMunichGermany
- German Center for Neurodegenerative Diseases (DZNE), site MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Jonathan Vöglein
- Department of NeurologyLudwig‐Maximilians‐Universität MünchenMunichGermany
- German Center for Neurodegenerative Diseases (DZNE), site MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - David M. Cash
- UK Dementia Research Institute at University College LondonLondonUK
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
| | - Igor Yakushev
- Department of NeurologyLudwig‐Maximilians‐Universität MünchenMunichGermany
- German Center for Neurodegenerative Diseases (DZNE), site MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | | | | | - John C. Morris
- Washington University School of MedicineSt. LouisMissouriUSA
| | | | | | | |
Collapse
|
21
|
Babulal GM, Chen L, Murphy SA, Doherty JM, Johnson AM, Morris JC. Neuropsychiatric Symptoms and Alzheimer Disease Biomarkers Independently Predict Progression to Incident Cognitive Impairment. Am J Geriatr Psychiatry 2023; 31:1190-1199. [PMID: 37544835 PMCID: PMC10861300 DOI: 10.1016/j.jagp.2023.07.012] [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: 06/13/2023] [Revised: 07/21/2023] [Accepted: 07/21/2023] [Indexed: 08/08/2023]
Abstract
OBJECTIVES To investigate the effect of neuropsychiatric symptoms and depression symptoms, respectively, and Alzheimer disease (AD) biomarkers (cerebrospinal fluid [CSF] or Positron Emission Tomography [PET] imaging) on the progression to incident cognitive impairment among cognitively normal older adults. DESIGN Prospective, observation, longitudinal study. SETTING Knight Alzheimer Disease Research Center (ADRC) at Washington University School of Medicine. PARTICIPANTS Older adults aged 65 and above who participated in AD longitudinal studies (n = 286). MEASUREMENTS CSF and PET biomarkers, Clinical Dementia Rating (CDR), Geriatric Depression Scale (GDS), and Neuropsychiatric Inventory Questionnaire (NPI-Q). RESULTS Participants had an average follow-up of eight years, and 31 progressed from CDR 0 to CDR >0. After adjusting for sex, age, and education in the Cox proportional hazards survival models, neuropsychiatric symptoms as a time-dependent covariate was statistically significant in the three CSF (Aβ42/Aβ40, t-Tau/Aβ42, p-Tau/Aβ42) PET imaging models (HR = 1.33-1.50). The biomarkers were also significant as main effects (HR = 2.00-4.04). Change in depression symptoms was not significant in any models. The interactions between biomarkers and neuropsychiatric symptoms and depression were not statistically significant. CONCLUSIONS Changes in neuropsychiatric symptoms increase the risk of progression to cognitive impairment among healthy, cognitively normal adults, independent of AD biomarkers. Routine assessment of neuropsychiatric symptoms could provide valuable clinical information about cognitive functioning and preclinical disease state.
Collapse
Affiliation(s)
- Ganesh M Babulal
- Department of Neurology (GMB, SAM, JCM), Washington University in St. Louis, St. Louis, MO; Institute of Public Health (GMB), Washington University in St. Louis, St. Louis, MO; Department of Psychology, Faculty of Humanities (GMB), University of Johannesburg, Johannesburg, South Africa; Department of Clinical Research and Leadership (GMB), The George Washington University School of Medicine and Health Sciences, Washington, DC.
| | - Ling Chen
- Division of Biostatistics (LC), Washington University in St. Louis, St. Louis, MO
| | - Samantha A Murphy
- Department of Neurology (GMB, SAM, JCM), Washington University in St. Louis, St. Louis, MO
| | - Jason M Doherty
- Department of Neurology (GMB, SAM, JCM), Washington University in St. Louis, St. Louis, MO
| | - Ann M Johnson
- Center for Clinical Studies (AMJ), Washington University in St. Louis, St. Louis, MO
| | - John C Morris
- Department of Neurology (GMB, SAM, JCM), Washington University in St. Louis, St. Louis, MO; Hope Center for Neurological Disorders (JCM), Washington University in St. Louis, St. Louis, MO
| |
Collapse
|
22
|
Li C, Liu M, Xia J, Mei L, Yang Q, Shi F, Zhang H, Shen D. Individualized Assessment of Brain Aβ Deposition With fMRI Using Deep Learning. IEEE J Biomed Health Inform 2023; 27:5430-5438. [PMID: 37616143 DOI: 10.1109/jbhi.2023.3306460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
PET-based Alzheimer's disease (AD) assessment has many limitations in large-scale screening. Non-invasive techniques such as resting-state functional magnetic resonance imaging (rs-fMRI) have been proven valuable in early AD diagnosis. This study investigated feasibility of using rs-fMRI, especially functional connectivity (FC), for individualized assessment of brain amyloid-β deposition derived from PET. We designed a graph convolutional networks (GCNs) and random forest (RF) based integrated framework for using rs-fMRI-derived multi-level FC networks to predict amyloid-β PET patterns with the OASIS-3 (N = 258) and ADNI-2 (N = 291) datasets. Our method achieved satisfactory accuracy not only in Aβ-PET grade classification (for negative, intermediate, and positive grades, with accuracy in the three-class classification as 62.8% and 64.3% on two datasets, respectively), but also in prediction of whole-brain region-level Aβ-PET standard uptake value ratios (SUVRs) (with the mean square errors as 0.039 and 0.074 for two datasets, respectively). Model interpretability examination also revealed the contributive role of the limbic network. This study demonstrated high feasibility and reproducibility of using low-cost, more accessible magnetic resonance imaging (MRI) to approximate PET-based diagnosis.
Collapse
|
23
|
Stojanovic M, Babulal GM, Head D. Determinants of physical activity engagement in older adults. J Behav Med 2023; 46:757-769. [PMID: 36920727 PMCID: PMC10502182 DOI: 10.1007/s10865-023-00404-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 02/21/2023] [Indexed: 03/16/2023]
Abstract
In order to increase engagement in physical activity, it is important to determine which factors contribute to physical activity engagement in older adults. The current study examined the relative predictive ability of several potential determinants, in terms of both the concurrent level as well as longitudinal trajectories. Clinically normal adults aged 61-92 completed the Physical Activity Scale for the Elderly (n = 189 for cross-sectional models; n = 214 for longitudinal models). Potential determinants included age, gender, education, physical health, sensory health, mood, cardiovascular health, cognitive status, and biomarkers of Alzheimer disease (AD). We observed a novel finding that both concurrent physical health (p < 0.001) and change in physical health (p < 0.001) were significant predictors above and beyond other determinants. Concurrent mood predicted levels of physical activity (p = 0.035), particularly in females. These findings suggest that poor physical health and low mood might be important to consider as potential barriers to physical activity engagement in older adults.
Collapse
Affiliation(s)
- Marta Stojanovic
- Department of Psychological & Brain Sciences, Washington University in St. Louis, One Brookings Drive, St. Louis, MO, Box 1125, USA.
| | - Ganesh M Babulal
- Department of Psychology, Faculty of Humanities, University of Johannesburg, Johannesburg, South Africa
- Department of Clinical Research and Leadership, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Institute of Public Health, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Denise Head
- Department of Psychological & Brain Sciences, Washington University in St. Louis, One Brookings Drive, St. Louis, MO, Box 1125, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
24
|
Wang L, Zheng Z, Su Y, Chen K, Weidman D, Wu T, Lo S, Lure F, Li J. Early Prediction of Progression to Alzheimer's Disease using Multi-Modality Neuroimages by a Novel Ordinal Learning Model ADPacer. IISE TRANSACTIONS ON HEALTHCARE SYSTEMS ENGINEERING 2023; 14:167-177. [PMID: 39239251 PMCID: PMC11374100 DOI: 10.1080/24725579.2023.2249487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Machine learning has shown great promise for integrating multi-modality neuroimaging datasets to predict the risk of progression/conversion to Alzheimer's Disease (AD) for individuals with Mild Cognitive Impairment (MCI). Most existing work aims to classify MCI patients into converters versus non-converters using a pre-defined timeframe. The limitation is a lack of granularity in differentiating MCI patients who convert at different paces. Progression pace prediction has important clinical values, which allow from more personalized interventional strategies, better preparation of patients and their caregivers, and facilitation of patient selection in clinical trials. We proposed a novel ADPacer model which formulated the pace prediction into an ordinal learning problem with a unique capability of leveraging training samples with label ambiguity to augment the training set. This capability differentiates ADPacer from existing ordinal learning algorithms. We applied ADPacer to MCI patient cohorts from the Alzheimer's Disease Neuroimaging Initiative (ADNI) and the Australian Imaging, Biomarker & Lifestyle Flagship Study of Ageing (AIBL), and demonstrated the superior performance of ADPacer compared to existing ordinal learning algorithms. We also integrated the SHapley Additive exPlanations (SHAP) method with ADPacer to assess the contributions from different modalities to the model prediction. The findings are consistent with the AD literature.
Collapse
Affiliation(s)
- Lujia Wang
- H. Hilton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, GA USA
| | - Zhiyang Zheng
- H. Hilton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, GA USA
| | - Yi Su
- Banner Alzheimer's Institute, AZ USA
| | | | | | - Teresa Wu
- School of Computing and Augmented Intelligence, Arizona State University, AZ USA
| | | | | | - Jing Li
- H. Hilton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, GA USA
| |
Collapse
|
25
|
McKay NS, Gordon BA, Hornbeck RC, Dincer A, Flores S, Keefe SJ, Joseph-Mathurin N, Jack CR, Koeppe R, Millar PR, Ances BM, Chen CD, Daniels A, Hobbs DA, Jackson K, Koudelis D, Massoumzadeh P, McCullough A, Nickels ML, Rahmani F, Swisher L, Wang Q, Allegri RF, Berman SB, Brickman AM, Brooks WS, Cash DM, Chhatwal JP, Day GS, Farlow MR, la Fougère C, Fox NC, Fulham M, Ghetti B, Graff-Radford N, Ikeuchi T, Klunk W, Lee JH, Levin J, Martins R, Masters CL, McConathy J, Mori H, Noble JM, Reischl G, Rowe C, Salloway S, Sanchez-Valle R, Schofield PR, Shimada H, Shoji M, Su Y, Suzuki K, Vöglein J, Yakushev I, Cruchaga C, Hassenstab J, Karch C, McDade E, Perrin RJ, Xiong C, Morris JC, Bateman RJ, Benzinger TLS. Positron emission tomography and magnetic resonance imaging methods and datasets within the Dominantly Inherited Alzheimer Network (DIAN). Nat Neurosci 2023; 26:1449-1460. [PMID: 37429916 PMCID: PMC10400428 DOI: 10.1038/s41593-023-01359-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/15/2023] [Indexed: 07/12/2023]
Abstract
The Dominantly Inherited Alzheimer Network (DIAN) is an international collaboration studying autosomal dominant Alzheimer disease (ADAD). ADAD arises from mutations occurring in three genes. Offspring from ADAD families have a 50% chance of inheriting their familial mutation, so non-carrier siblings can be recruited for comparisons in case-control studies. The age of onset in ADAD is highly predictable within families, allowing researchers to estimate an individual's point in the disease trajectory. These characteristics allow candidate AD biomarker measurements to be reliably mapped during the preclinical phase. Although ADAD represents a small proportion of AD cases, understanding neuroimaging-based changes that occur during the preclinical period may provide insight into early disease stages of 'sporadic' AD also. Additionally, this study provides rich data for research in healthy aging through inclusion of the non-carrier controls. Here we introduce the neuroimaging dataset collected and describe how this resource can be used by a range of researchers.
Collapse
Affiliation(s)
| | | | | | - Aylin Dincer
- Washington University in St. Louis, St. Louis, MO, USA
| | - Shaney Flores
- Washington University in St. Louis, St. Louis, MO, USA
| | - Sarah J Keefe
- Washington University in St. Louis, St. Louis, MO, USA
| | | | | | | | | | - Beau M Ances
- Washington University in St. Louis, St. Louis, MO, USA
| | | | | | - Diana A Hobbs
- Washington University in St. Louis, St. Louis, MO, USA
| | | | | | | | | | | | | | - Laura Swisher
- Washington University in St. Louis, St. Louis, MO, USA
| | - Qing Wang
- Washington University in St. Louis, St. Louis, MO, USA
| | | | | | - Adam M Brickman
- Columbia University Irving Medical Center, New York, NY, USA
| | - William S Brooks
- Neuroscience Research Australia, Sydney, New South Wales, Australia
| | - David M Cash
- UK Dementia Research Institute at University College London, London, UK
- University College London, London, UK
| | - Jasmeer P Chhatwal
- Massachusetts General and Brigham & Women's Hospitals, Harvard Medical School, Boston, MA, USA
| | | | | | - Christian la Fougère
- Department of Radiology, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Nick C Fox
- UK Dementia Research Institute at University College London, London, UK
- University College London, London, UK
| | - Michael Fulham
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | | | | | | | | | | | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Ralph Martins
- Edith Cowan University, Joondalup, Western Australia, Australia
| | | | | | | | - James M Noble
- Columbia University Irving Medical Center, New York, NY, USA
| | - Gerald Reischl
- Department of Radiology, University of Tübingen, Tübingen, Germany
| | | | | | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, New South Wales, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | | | | | - Yi Su
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | | | - Jonathan Vöglein
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Ludwig-Maximilians-Universität München, München, Germany
| | - Igor Yakushev
- School of Medicine, Technical University of Munich, Munich, Germany
| | | | | | - Celeste Karch
- Washington University in St. Louis, St. Louis, MO, USA
| | - Eric McDade
- Washington University in St. Louis, St. Louis, MO, USA
| | | | | | - John C Morris
- Washington University in St. Louis, St. Louis, MO, USA
| | | | | |
Collapse
|
26
|
Aschenbrenner AJ, Crawford JL, Peelle JE, Fagan AM, Benzinger TLS, Morris JC, Hassenstab J, Braver TS. Increased cognitive effort costs in healthy aging and preclinical Alzheimer's disease. Psychol Aging 2023; 38:428-442. [PMID: 37067479 PMCID: PMC10440282 DOI: 10.1037/pag0000742] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Life-long engagement in cognitively demanding activities may mitigate against declines in cognitive ability observed in healthy or pathological aging. However, the "mental costs" associated with completing cognitive tasks also increase with age and may be partly attributed to increases in preclinical levels of Alzheimer's disease (AD) pathology, specifically amyloid. We test whether cognitive effort costs increase in a domain-general manner among older adults, and further, whether such age-related increases in cognitive effort costs are associated with working memory (WM) capacity or amyloid burden, a signature pathology of AD. In two experiments, we administered a behavioral measure of cognitive effort costs (cognitive effort discounting) to a sample of older adults recruited from online sources (Experiment 1) or from ongoing longitudinal studies of aging and dementia (Experiment 2). Experiment 1 compared age-related differences in cognitive effort costs across two domains, WM and speech comprehension. Experiment 2 compared cognitive effort costs between a group of participants who were rated positive for amyloid relative to those with no evidence of amyloid. Results showed age-related increases in cognitive effort costs were evident in both domains. Cost estimates were highly correlated between the WM and speech comprehension tasks but did not correlate with WM capacity. In addition, older adults who were amyloid positive had higher cognitive effort costs than those who were amyloid negative. Cognitive effort costs may index a domain-general trait that consistently increases in aging. Differences in cognitive effort costs associated with amyloid burden suggest a potential neurobiological mechanism for age-related differences. (PsycInfo Database Record (c) 2023 APA, all rights reserved).
Collapse
Affiliation(s)
| | - Jennifer L Crawford
- Department of Psychological and Brain Sciences, Washington University in St. Louis
| | | | - Anne M Fagan
- Department of Neurology, Washington University in St. Louis
| | | | - John C Morris
- Department of Neurology, Washington University in St. Louis
| | | | - Todd S Braver
- Department of Psychological and Brain Sciences, Washington University in St. Louis
| |
Collapse
|
27
|
Schultz SA, Shirzadi Z, Schultz AP, Liu L, Fitzpatrick CD, McDade E, Barthelemy NR, Renton A, Esposito B, Joseph‐Mathurin N, Cruchaga C, Chen CD, Goate A, Allegri RF, Benzinger TLS, Berman S, Chui HC, Fagan AM, Farlow MR, Fox NC, Gordon BA, Day GS, Graff‐Radford NR, Hassenstab JJ, Hanseeuw BJ, Hofmann A, Jack CR, Jucker M, Karch CM, Koeppe RA, Lee J, Levey AI, Levin J, Martins RN, Mori H, Morris JC, Noble J, Perrin RJ, Rosa‐Neto P, Salloway SP, Sanchez‐Valle R, Schofield PR, Xiong C, Johnson KA, Bateman RJ, Sperling RA, Chhatwal JP. Location of pathogenic variants in PSEN1 impacts progression of cognitive, clinical, and neurodegenerative measures in autosomal-dominant Alzheimer's disease. Aging Cell 2023; 22:e13871. [PMID: 37291760 PMCID: PMC10410059 DOI: 10.1111/acel.13871] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/30/2023] [Accepted: 05/03/2023] [Indexed: 06/10/2023] Open
Abstract
Although pathogenic variants in PSEN1 leading to autosomal-dominant Alzheimer disease (ADAD) are highly penetrant, substantial interindividual variability in the rates of cognitive decline and biomarker change are observed in ADAD. We hypothesized that this interindividual variability may be associated with the location of the pathogenic variant within PSEN1. PSEN1 pathogenic variant carriers participating in the Dominantly Inherited Alzheimer Network (DIAN) observational study were grouped based on whether the underlying variant affects a transmembrane (TM) or cytoplasmic (CY) protein domain within PSEN1. CY and TM carriers and variant non-carriers (NC) who completed clinical evaluation, multimodal neuroimaging, and lumbar puncture for collection of cerebrospinal fluid (CSF) as part of their participation in DIAN were included in this study. Linear mixed effects models were used to determine differences in clinical, cognitive, and biomarker measures between the NC, TM, and CY groups. While both the CY and TM groups were found to have similarly elevated Aβ compared to NC, TM carriers had greater cognitive impairment, smaller hippocampal volume, and elevated phosphorylated tau levels across the spectrum of pre-symptomatic and symptomatic phases of disease as compared to CY, using both cross-sectional and longitudinal data. As distinct portions of PSEN1 are differentially involved in APP processing by γ-secretase and the generation of toxic β-amyloid species, these results have important implications for understanding the pathobiology of ADAD and accounting for a substantial portion of the interindividual heterogeneity in ongoing ADAD clinical trials.
Collapse
Affiliation(s)
| | - Zahra Shirzadi
- Massachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Aaron P. Schultz
- Massachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Lei Liu
- Brigham and Women's HospitalBostonMassachusettsUSA
- Ann Romney Center for Neurologic DiseasesBostonMassachusettsUSA
| | | | - Eric McDade
- Washington University in St. Louis School of MedicineSt. LouisMissouriUSA
| | | | - Alan Renton
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Bianca Esposito
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | | | - Carlos Cruchaga
- Washington University in St. Louis School of MedicineSt. LouisMissouriUSA
| | - Charles D. Chen
- Washington University in St. Louis School of MedicineSt. LouisMissouriUSA
| | - Alison Goate
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | | | | | - Sarah Berman
- University of PittsburghPittsburghPennsylvaniaUSA
| | - Helena C. Chui
- Department of Neurology, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Anne M. Fagan
- Washington University in St. Louis School of MedicineSt. LouisMissouriUSA
| | - Martin R. Farlow
- Indiana Alzheimer's Disease Research CenterIndianapolisIndianaUSA
| | - Nick C. Fox
- Dementia Research Centre & UK Dementia Research InstituteUCL Institute of NeurologyLondonUK
| | - Brian A. Gordon
- Washington University in St. Louis School of MedicineSt. LouisMissouriUSA
| | | | | | | | - Bernard J. Hanseeuw
- Institute of Neuroscience, UCLouvainBrusselsBelgium
- Gordon Center for Medical Imaging in the Radiology Department of MGHBostonMassachusettsUSA
| | - Anna Hofmann
- German Center for Neurodegenerative Diseases (DZNE)TuebingenGermany
| | | | - Mathias Jucker
- German Center for Neurodegenerative Diseases (DZNE)TuebingenGermany
| | - Celeste M. Karch
- Washington University in St. Louis School of MedicineSt. LouisMissouriUSA
| | | | - Jae‐Hong Lee
- Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Allan I. Levey
- Emory Goizueta Alzheimer's Disease Research CenterAtlantaGeorgiaUSA
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
- Department of NeurologyLudwig‐Maximilians‐Universität MünchenMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | | | | | - John C. Morris
- Washington University in St. Louis School of MedicineSt. LouisMissouriUSA
| | | | - Richard J. Perrin
- Washington University in St. Louis School of MedicineSt. LouisMissouriUSA
| | - Pedro Rosa‐Neto
- Translational Neuroimaging Laboratory, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest‐de‐l'Île‐de‐Montréal; Department of Neurology and NeurosurgeryMcGill UniversityMontrealCanada
| | | | - Raquel Sanchez‐Valle
- Alzheimer's disease and other cognitive disorders Unit, Neurology Department, Hospital Clínic de BarcelonaInstitut d'Investigacions BiomediquesBarcelonaSpain
| | - Peter R. Schofield
- Neuroscience Research AustraliaRandwickNew South WalesAustralia
- School of Medical SciencesUniversity of New South WalesSydneyNew South WalesAustralia
| | - Chengjie Xiong
- Washington University in St. Louis School of MedicineSt. LouisMissouriUSA
| | - Keith A. Johnson
- Massachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Brigham and Women's HospitalBostonMassachusettsUSA
| | - Randall J. Bateman
- Washington University in St. Louis School of MedicineSt. LouisMissouriUSA
| | - Reisa A. Sperling
- Massachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Brigham and Women's HospitalBostonMassachusettsUSA
| | - Jasmeer P. Chhatwal
- Massachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Brigham and Women's HospitalBostonMassachusettsUSA
| | | |
Collapse
|
28
|
Wisch JK, Butt OH, Gordon BA, Schindler SE, Fagan AM, Henson RL, Yang C, Boerwinkle AH, Benzinger TLS, Holtzman DM, Morris JC, Cruchaga C, Ances BM. Proteomic clusters underlie heterogeneity in preclinical Alzheimer's disease progression. Brain 2023; 146:2944-2956. [PMID: 36542469 PMCID: PMC10316757 DOI: 10.1093/brain/awac484] [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: 04/05/2022] [Revised: 11/21/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
Heterogeneity in progression to Alzheimer's disease (AD) poses challenges for both clinical prognosis and clinical trial implementation. Multiple AD-related subtypes have previously been identified, suggesting differences in receptivity to drug interventions. We identified early differences in preclinical AD biomarkers, assessed patterns for developing preclinical AD across the amyloid-tau-(neurodegeneration) [AT(N)] framework, and considered potential sources of difference by analysing the CSF proteome. Participants (n = 10) enrolled in longitudinal studies at the Knight Alzheimer Disease Research Center completed four or more lumbar punctures. These individuals were cognitively normal at baseline. Cerebrospinal fluid measures of amyloid-β (Aβ)42, phosphorylated tau (pTau181), and neurofilament light chain (NfL) as well as proteomics values were evaluated. Imaging biomarkers, including PET amyloid and tau, and structural MRI, were repeatedly obtained when available. Individuals were staged according to the amyloid-tau-(neurodegeneration) framework. Growth mixture modelling, an unsupervised clustering technique, identified three patterns of biomarker progression as measured by CSF pTau181 and Aβ42. Two groups (AD Biomarker Positive and Intermediate AD Biomarker) showed distinct progression from normal biomarker status to having biomarkers consistent with preclinical AD. A third group (AD Biomarker Negative) did not develop abnormal AD biomarkers over time. Participants grouped by CSF trajectories were re-classified using only proteomic profiles (AUCAD Biomarker Positive versus AD Biomarker Negative = 0.857, AUCAD Biomarker Positive versus Intermediate AD Biomarkers = 0.525, AUCIntermediate AD Biomarkers versus AD Biomarker Negative = 0.952). We highlight heterogeneity in the development of AD biomarkers in cognitively normal individuals. We identified some individuals who became amyloid positive before the age of 50 years. A second group, Intermediate AD Biomarkers, developed elevated CSF ptau181 significantly before becoming amyloid positive. A third group were AD Biomarker Negative over repeated testing. Our results could influence the selection of participants for specific treatments (e.g. amyloid-reducing versus other agents) in clinical trials. CSF proteome analysis highlighted additional non-AT(N) biomarkers for potential therapies, including blood-brain barrier-, vascular-, immune-, and neuroinflammatory-related targets.
Collapse
Affiliation(s)
- Julie K Wisch
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Omar H Butt
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Brian A Gordon
- Department of Radiology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Hope Center, Washington University in Saint Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anne M Fagan
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rachel L Henson
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Chengran Yang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anna H Boerwinkle
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Hope Center, Washington University in Saint Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John C Morris
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carlos Cruchaga
- Hope Center, Washington University in Saint Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Beau M Ances
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Hope Center, Washington University in Saint Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
29
|
Wheelock MD, Strain JF, Mansfield P, Tu JC, Tanenbaum A, Preische O, Chhatwal JP, Cash DM, Cruchaga C, Fagan AM, Fox NC, Graff-Radford NR, Hassenstab J, Jack CR, Karch CM, Levin J, McDade EM, Perrin RJ, Schofield PR, Xiong C, Morris JC, Bateman RJ, Jucker M, Benzinger TLS, Ances BM, Eggebrecht AT, Gordon BA, Allegri R, Araki A, Barthelemy N, Bateman R, Bechara J, Benzinger T, Berman S, Bodge C, Brandon S, Brooks W, Brosch J, Buck J, Buckles V, Carter K, Cash D, Cash L, Chen C, Chhatwal J, Chrem P, Chua J, Chui H, Cruchaga C, Day GS, De La Cruz C, Denner D, Diffenbacher A, Dincer A, Donahue T, Douglas J, Duong D, Egido N, Esposito B, Fagan A, Farlow M, Feldman B, Fitzpatrick C, Flores S, Fox N, Franklin E, Friedrichsen N, Fujii H, Gardener S, Ghetti B, Goate A, Goldberg S, Goldman J, Gonzalez A, Gordon B, Gräber-Sultan S, Graff-Radford N, Graham M, Gray J, Gremminger E, Grilo M, Groves A, Haass C, Häsler L, Hassenstab J, Hellm C, Herries E, Hoechst-Swisher L, Hofmann A, Holtzman D, Hornbeck R, Igor Y, Ihara R, Ikeuchi T, Ikonomovic S, Ishii K, Jack C, Jerome G, Johnson E, Jucker M, Karch C, Käser S, Kasuga K, Keefe S, Klunk W, Koeppe R, Koudelis D, Kuder-Buletta E, Laske C, Lee JH, Levey A, Levin J, Li Y, Lopez O, Marsh J, Martinez R, Martins R, Mason NS, Masters C, Mawuenyega K, McCullough A, McDade E, Mejia A, Morenas-Rodriguez E, Mori H, Morris J, Mountz J, Mummery C, Nadkami N, Nagamatsu A, Neimeyer K, Niimi Y, Noble J, Norton J, Nuscher B, O'Connor A, Obermüller U, Patira R, Perrin R, Ping L, Preische O, Renton A, Ringman J, Salloway S, Sanchez-Valle R, Schofield P, Senda M, Seyfried N, Shady K, Shimada H, Sigurdson W, Smith J, Smith L, Snitz B, Sohrabi H, Stephens S, Taddei K, Thompson S, Vöglein J, Wang P, Wang Q, Weamer E, Xiong C, Xu J, Xu X. Brain network decoupling with increased serum neurofilament and reduced cognitive function in Alzheimer's disease. Brain 2023; 146:2928-2943. [PMID: 36625756 PMCID: PMC10316768 DOI: 10.1093/brain/awac498] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 11/21/2022] [Accepted: 12/12/2022] [Indexed: 01/11/2023] Open
Abstract
Neurofilament light chain, a putative measure of neuronal damage, is measurable in blood and CSF and is predictive of cognitive function in individuals with Alzheimer's disease. There has been limited prior work linking neurofilament light and functional connectivity, and no prior work has investigated neurofilament light associations with functional connectivity in autosomal dominant Alzheimer's disease. Here, we assessed relationships between blood neurofilament light, cognition, and functional connectivity in a cross-sectional sample of 106 autosomal dominant Alzheimer's disease mutation carriers and 76 non-carriers. We employed an innovative network-level enrichment analysis approach to assess connectome-wide associations with neurofilament light. Neurofilament light was positively correlated with deterioration of functional connectivity within the default mode network and negatively correlated with connectivity between default mode network and executive control networks, including the cingulo-opercular, salience, and dorsal attention networks. Further, reduced connectivity within the default mode network and between the default mode network and executive control networks was associated with reduced cognitive function. Hierarchical regression analysis revealed that neurofilament levels and functional connectivity within the default mode network and between the default mode network and the dorsal attention network explained significant variance in cognitive composite scores when controlling for age, sex, and education. A mediation analysis demonstrated that functional connectivity within the default mode network and between the default mode network and dorsal attention network partially mediated the relationship between blood neurofilament light levels and cognitive function. Our novel results indicate that blood estimates of neurofilament levels correspond to direct measurements of brain dysfunction, shedding new light on the underlying biological processes of Alzheimer's disease. Further, we demonstrate how variation within key brain systems can partially mediate the negative effects of heightened total serum neurofilament levels, suggesting potential regions for targeted interventions. Finally, our results lend further evidence that low-cost and minimally invasive blood measurements of neurofilament may be a useful marker of brain functional connectivity and cognitive decline in Alzheimer's disease.
Collapse
Affiliation(s)
- Muriah D Wheelock
- Department of Radiology, Washington University in St. Louis, MO, USA
| | - Jeremy F Strain
- Department of Neurology, Washington University in Saint Louis, St. Louis, MO, USA
| | | | - Jiaxin Cindy Tu
- Department of Radiology, Washington University in St. Louis, MO, USA
| | - Aaron Tanenbaum
- Department of Neurology, Washington University in Saint Louis, St. Louis, MO, USA
| | - Oliver Preische
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - David M Cash
- Dementia Research Center, UCL Queen Square, London, UK.,UK Dementia Research Institute, College London, London, UK
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St. Louis, MO, USA
| | - Anne M Fagan
- Department of Neurology, Washington University in Saint Louis, St. Louis, MO, USA
| | - Nick C Fox
- Dementia Research Center, UCL Queen Square, London, UK.,UK Dementia Research Institute, College London, London, UK
| | | | - Jason Hassenstab
- Department of Neurology, Washington University in Saint Louis, St. Louis, MO, USA
| | | | - Celeste M Karch
- Department of Psychiatry, Washington University in St. Louis, MO, USA
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Eric M McDade
- Department of Neurology, Washington University in Saint Louis, St. Louis, MO, USA
| | - Richard J Perrin
- Department of Neurology, Washington University in Saint Louis, St. Louis, MO, USA.,Department of Pathology & Immunology, Washington University in St. Louis, MO, USA
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, NSW, Australia.,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Chengjie Xiong
- Division of Biostatistics, Washington University in St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University in Saint Louis, St. Louis, MO, USA
| | - Randal J Bateman
- Department of Neurology, Washington University in Saint Louis, St. Louis, MO, USA
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Tammie L S Benzinger
- Department of Neurology, Washington University in Saint Louis, St. Louis, MO, USA
| | - Beau M Ances
- Department of Neurology, Washington University in Saint Louis, St. Louis, MO, USA
| | - Adam T Eggebrecht
- Department of Radiology, Washington University in St. Louis, MO, USA
| | - Brian A Gordon
- Department of Radiology, Washington University in St. Louis, MO, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Chen CD, McCullough A, Gordon B, Joseph-Mathurin N, Flores S, McKay NS, Hobbs DA, Hornbeck R, Fagan AM, Cruchaga C, Goate AM, Perrin RJ, Wang G, Li Y, Shi X, Xiong C, Pontecorvo MJ, Klein G, Su Y, Klunk WE, Jack C, Koeppe R, Snider BJ, Berman SB, Roberson ED, Brosch J, Surti G, Jiménez-Velázquez IZ, Galasko D, Honig LS, Brooks WS, Clarnette R, Wallon D, Dubois B, Pariente J, Pasquier F, Sanchez-Valle R, Shcherbinin S, Higgins I, Tunali I, Masters CL, van Dyck CH, Masellis M, Hsiung R, Gauthier S, Salloway S, Clifford DB, Mills S, Supnet-Bell C, McDade E, Bateman RJ, Benzinger TLS. Longitudinal head-to-head comparison of 11C-PiB and 18F-florbetapir PET in a Phase 2/3 clinical trial of anti-amyloid-β monoclonal antibodies in dominantly inherited Alzheimer's disease. Eur J Nucl Med Mol Imaging 2023; 50:2669-2682. [PMID: 37017737 PMCID: PMC10330155 DOI: 10.1007/s00259-023-06209-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/18/2023] [Indexed: 04/06/2023]
Abstract
PURPOSE Pittsburgh Compound-B (11C-PiB) and 18F-florbetapir are amyloid-β (Aβ) positron emission tomography (PET) radiotracers that have been used as endpoints in Alzheimer's disease (AD) clinical trials to evaluate the efficacy of anti-Aβ monoclonal antibodies. However, comparing drug effects between and within trials may become complicated if different Aβ radiotracers were used. To study the consequences of using different Aβ radiotracers to measure Aβ clearance, we performed a head-to-head comparison of 11C-PiB and 18F-florbetapir in a Phase 2/3 clinical trial of anti-Aβ monoclonal antibodies. METHODS Sixty-six mutation-positive participants enrolled in the gantenerumab and placebo arms of the first Dominantly Inherited Alzheimer Network Trials Unit clinical trial (DIAN-TU-001) underwent both 11C-PiB and 18F-florbetapir PET imaging at baseline and during at least one follow-up visit. For each PET scan, regional standardized uptake value ratios (SUVRs), regional Centiloids, a global cortical SUVR, and a global cortical Centiloid value were calculated. Longitudinal changes in SUVRs and Centiloids were estimated using linear mixed models. Differences in longitudinal change between PET radiotracers and between drug arms were estimated using paired and Welch two sample t-tests, respectively. Simulated clinical trials were conducted to evaluate the consequences of some research sites using 11C-PiB while other sites use 18F-florbetapir for Aβ PET imaging. RESULTS In the placebo arm, the absolute rate of longitudinal change measured by global cortical 11C-PiB SUVRs did not differ from that of global cortical 18F-florbetapir SUVRs. In the gantenerumab arm, global cortical 11C-PiB SUVRs decreased more rapidly than global cortical 18F-florbetapir SUVRs. Drug effects were statistically significant across both Aβ radiotracers. In contrast, the rates of longitudinal change measured in global cortical Centiloids did not differ between Aβ radiotracers in either the placebo or gantenerumab arms, and drug effects remained statistically significant. Regional analyses largely recapitulated these global cortical analyses. Across simulated clinical trials, type I error was higher in trials where both Aβ radiotracers were used versus trials where only one Aβ radiotracer was used. Power was lower in trials where 18F-florbetapir was primarily used versus trials where 11C-PiB was primarily used. CONCLUSION Gantenerumab treatment induces longitudinal changes in Aβ PET, and the absolute rates of these longitudinal changes differ significantly between Aβ radiotracers. These differences were not seen in the placebo arm, suggesting that Aβ-clearing treatments may pose unique challenges when attempting to compare longitudinal results across different Aβ radiotracers. Our results suggest converting Aβ PET SUVR measurements to Centiloids (both globally and regionally) can harmonize these differences without losing sensitivity to drug effects. Nonetheless, until consensus is achieved on how to harmonize drug effects across radiotracers, and since using multiple radiotracers in the same trial may increase type I error, multisite studies should consider potential variability due to different radiotracers when interpreting Aβ PET biomarker data and, if feasible, use a single radiotracer for the best results. TRIAL REGISTRATION ClinicalTrials.gov NCT01760005. Registered 31 December 2012. Retrospectively registered.
Collapse
Affiliation(s)
- Charles D Chen
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
- Washington University School of Medicine, 660 South Euclid, Campus Box 8225, St. Louis, MO, 63110, USA
| | - Austin McCullough
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Brian Gordon
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Nelly Joseph-Mathurin
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Shaney Flores
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Nicole S McKay
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Diana A Hobbs
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Russ Hornbeck
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Anne M Fagan
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Alison M Goate
- Department of Genetics and Genomic Sciences, Ichan School of Medicine at Mount Sinai, New York, NY, USA
| | - Richard J Perrin
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA
| | - Guoqiao Wang
- Department of Biostatistics, Washington University in St. Louis, St. Louis, MO, USA
| | - Yan Li
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Xinyu Shi
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Chengjie Xiong
- Department of Biostatistics, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael J Pontecorvo
- Avid Radiopharmaceuticals, Philadelphia, PA, USA
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Yi Su
- Banner Alzheimer's Institute, Banner Health, Phoenix, AZ, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - William E Klunk
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Clifford Jack
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Robert Koeppe
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - B Joy Snider
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Sarah B Berman
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Erik D Roberson
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jared Brosch
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ghulam Surti
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | | | - Douglas Galasko
- Department of Neurology, University of California San Diego, San Diego, CA, USA
| | | | - William S Brooks
- Prince of Wales Medical Research Institute, University of New South Wales, Sydney, NSW, Australia
| | - Roger Clarnette
- Department of Internal Medicine, University of Western Australia, Crawley, WA, Australia
| | - David Wallon
- Department of Neurology and CNR-MAJ, Normandie Univ, UNIROUEN, INSERM U1245, CHU Rouen, F-76000, Rouen, France
| | - Bruno Dubois
- Sorbonne Université, AP-HP, GRC No. 21, APM, Hôpital de La Pitié-Salpêtrière, Paris, France
- Institut du Cerveau Et de La Moelle Épinière, INSERM U1127, CNRS UMR 7225, Paris, France
- Institut de La Mémoire Et de La Maladie d'Alzheimer, Département de Neurologie, Hôpital de La Pitié-Salpêtrière, Paris, France
| | - Jérémie Pariente
- Department of Neurology, Hôpital Pierre-Paul Riquet, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
- Toulouse NeuroImaging Centre, Université de Toulouse, INSERM, UPS, Toulouse, France
| | - Florence Pasquier
- Univ. Lille, INSERM, CHU Lille, 59000, Lille, France
- CNR-MAJ, Labex DISTALZ, LiCEND, 59000, Lille, France
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Hospital ClínicInstitut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Fundació Clínic Per a La Recerca Biomèdica, University of Barcelona, Barcelona, Spain
| | | | | | - Ilke Tunali
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | | | | | - Robin Hsiung
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Serge Gauthier
- Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Steve Salloway
- Alpert Medical School of Brown University, Providence, RI, USA
| | - David B Clifford
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Susan Mills
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Eric McDade
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Randall J Bateman
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
31
|
Xiong C, McCue LM, Buckles V, Grant E, Agboola F, Coble D, Bateman RJ, Fagan AM, Benzinger TL, Hassenstab J, Schindler SE, McDade E, Moulder K, Gordon BA, Cruchaga C, Day GS, Ikeuchi T, Suzuki K, Allegri RF, Vöglein J, Levin J, Morris JC. Cross-sectional and longitudinal comparisons of biomarkers and cognition among asymptomatic middle-aged individuals with a parental history of either autosomal dominant or late-onset Alzheimer's disease. Alzheimers Dement 2023; 19:2923-2932. [PMID: 36640138 PMCID: PMC10345163 DOI: 10.1002/alz.12912] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 01/15/2023]
Abstract
BACKGROUND Comparisons of late-onset Alzheimer's disease (LOAD) and autosomal dominant AD (ADAD) are confounded by age. METHODS We compared biomarkers from cerebrospinal fluid (CSF), magnetic resonance imaging, and amyloid imaging with Pittsburgh Compound-B (PiB) across four groups of 387 cognitively normal participants, 42 to 65 years of age, in the Dominantly Inherited Alzheimer Network (DIAN) and the Adult Children Study (ACS) of LOAD: DIAN mutation carriers (MCs) and non-carriers (NON-MCs), and ACS participants with a positive (FH+) and negative (FH-) family history of LOAD. RESULTS At baseline, MCs had the lowest age-adjusted level of CSF Aβ42 and the highest levels of total and phosphorylated tau-181, and PiB uptake. Longitudinally, MC had similar increase in PiB uptake to FH+, but drastically faster decline in hippocampal volume than others, and was the only group showing cognitive decline. DISCUSSION Preclinical ADAD and LOAD share many biomarker signatures, but cross-sectional and longitudinal differences may exist.
Collapse
Affiliation(s)
- Chengjie Xiong
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Neurology, Washington University, St. Louis, Missouri, USA
- Division of Biostatistics, Washington University, St. Louis, Missouri, USA
| | - Lena M. McCue
- Division of Biostatistics, Washington University, St. Louis, Missouri, USA
| | - Virginia Buckles
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Neurology, Washington University, St. Louis, Missouri, USA
| | - Elizabeth Grant
- Division of Biostatistics, Washington University, St. Louis, Missouri, USA
| | - Folasade Agboola
- Division of Biostatistics, Washington University, St. Louis, Missouri, USA
| | - Dean Coble
- Division of Biostatistics, Washington University, St. Louis, Missouri, USA
| | - Randall J. Bateman
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Neurology, Washington University, St. Louis, Missouri, USA
| | - Anne M Fagan
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Neurology, Washington University, St. Louis, Missouri, USA
| | - Tammie L.S. Benzinger
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Radiology, Washington University, St. Louis, Missouri, USA
- Department of Neurological Surgery, Washington University, St. Louis, Missouri, USA
| | - Jason Hassenstab
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Neurology, Washington University, St. Louis, Missouri, USA
- Department of Psychology, Washington University, St. Louis, Missouri, USA
| | - Suzanne E. Schindler
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Neurology, Washington University, St. Louis, Missouri, USA
| | - Eric McDade
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Neurology, Washington University, St. Louis, Missouri, USA
| | - Krista Moulder
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Neurology, Washington University, St. Louis, Missouri, USA
| | - Brian A. Gordon
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Psychology, Washington University, St. Louis, Missouri, USA
- Department of Radiology, Washington University, St. Louis, Missouri, USA
| | - Carlos Cruchaga
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- Department of Psychiatry, Washington University, St. Louis, Missouri, USA
| | - Gregory S. Day
- Department of Neurology, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, JAPAN
| | | | | | - Jonathan Vöglein
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - John C. Morris
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Neurology, Washington University, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri, USA
- Department of Physical Therapy, Washington University, St. Louis, Missouri, USA
- Department of Occupational Therapy, Washington University, St. Louis, Missouri, USA
| | | |
Collapse
|
32
|
Chatterjee P, Vermunt L, Gordon BA, Pedrini S, Boonkamp L, Armstrong NJ, Xiong C, Singh AK, Li Y, Sohrabi HR, Taddei K, Molloy MP, Benzinger TL, Morris JC, Karch CM, Berman SB, Chhatwal J, Cruchaga C, Graff-Radford NR, Day GS, Farlow M, Fox NC, Goate AM, Hassenstab J, Lee JH, Levin J, McDade E, Mori H, Perrin RJ, Sanchez-Valle R, Schofield PR, Levey A, Jucker M, Masters CL, Fagan AM, Bateman RJ, Martins RN, Teunissen CE. Plasma glial fibrillary acidic protein in autosomal dominant Alzheimer's disease: Associations with Aβ-PET, neurodegeneration, and cognition. Alzheimers Dement 2023; 19:2790-2804. [PMID: 36576155 PMCID: PMC10300233 DOI: 10.1002/alz.12879] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/22/2022] [Accepted: 10/21/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND Glial fibrillary acidic protein (GFAP) is a promising candidate blood-based biomarker for Alzheimer's disease (AD) diagnosis and prognostication. The timing of its disease-associated changes, its clinical correlates, and biofluid-type dependency will influence its clinical utility. METHODS We evaluated plasma, serum, and cerebrospinal fluid (CSF) GFAP in families with autosomal dominant AD (ADAD), leveraging the predictable age at symptom onset to determine changes by stage of disease. RESULTS Plasma GFAP elevations appear a decade before expected symptom onset, after amyloid beta (Aβ) accumulation and prior to neurodegeneration and cognitive decline. Plasma GFAP distinguished Aβ-positive from Aβ-negative ADAD participants and showed a stronger relationship with Aβ load in asymptomatic than symptomatic ADAD. Higher plasma GFAP was associated with the degree and rate of neurodegeneration and cognitive impairment. Serum GFAP showed similar relationships, but these were less pronounced for CSF GFAP. CONCLUSION Our findings support a role for plasma GFAP as a clinical biomarker of Aβ-related astrocyte reactivity that is associated with cognitive decline and neurodegeneration. HIGHLIGHTS Plasma glial fibrillary acidic protein (GFAP) elevations appear a decade before expected symptom onset in autosomal dominant Alzheimer's disease (ADAD). Plasma GFAP was associated to amyloid positivity in asymptomatic ADAD. Plasma GFAP increased with clinical severity and predicted disease progression. Plasma and serum GFAP carried similar information in ADAD, while cerebrospinal fluid GFAP did not.
Collapse
Affiliation(s)
- Pratishtha Chatterjee
- Macquarie Medical School, Macquarie University, North Ryde, NSW 2019, Australia; School of Medical Sciences, Edith Cowan University, Sarich Neuroscience Research Institute, Nedlands, WA 6009, Australia
| | - Lisa Vermunt
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, programme Neurodegeneration, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, The Netherlands
| | - Brian A. Gordon
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Steve Pedrini
- School of Medical Sciences, Edith Cowan University, Sarich Neuroscience Research Institute, Nedlands, WA 6009, Australia
| | - Lynn Boonkamp
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, programme Neurodegeneration, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, The Netherlands
| | - Nicola J. Armstrong
- Department of Mathematics & Statistics, Curtin University, Bentley, WA, Australia
| | - Chengjie Xiong
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA; Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Abhay K. Singh
- Macquarie Business School, Macquarie University, North Ryde, NSW, Australia
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hamid R. Sohrabi
- Department of Biomedical Sciences, Macquarie University, North Ryde, NSW 2019, Australia; School of Medical Sciences, Edith Cowan University, Sarich Neuroscience Research Institute, Nedlands, WA 6009, Australia; School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, WA, Australia; Australian Alzheimer’s Research Foundation, Nedlands, WA, Australia; Centre for Healthy Ageing, Health Future Institute, Murdoch University, Murdoch, WA, Australia
| | - Kevin Taddei
- School of Medical Sciences, Edith Cowan University, Sarich Neuroscience Research Institute, Nedlands, WA 6009, Australia; Australian Alzheimer’s Research Foundation, Nedlands, WA, Australia
| | - Mark P. Molloy
- Bowel Cancer and Biomarker Laboratory, Kolling Institute, The University of Sydney, St Leonards, NSW, Australia; Australian Proteome Analysis Facility, Macquarie University, North Ryde, NSW, Australia
| | - Tammie L.S. Benzinger
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - John C. Morris
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA; Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Celeste M. Karch
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Sarah B. Berman
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jasmeer Chhatwal
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Carlos Cruchaga
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Gregory S Day
- Department of Neurology, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - Martin Farlow
- Department of Neurology, Indiana University, Indianapolis, IN, USA
| | - Nick C. Fox
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK
| | - Alison M. Goate
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jae-Hong Lee
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul05505, Republic of Korea
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Eric McDade
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hiroshi Mori
- Osaka Metropolitan University, Nagaoka Sutoku University, Osaka, Japan
| | - Richard J. Perrin
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA; Dominantly Inherited Alzheimer Network, Washington University School of Medicine, St. Louis, MO, USA; Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA; Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Raquel Sanchez-Valle
- Alzheimer’s Disease and other Cognitive Disorders Unit, Neurology Service, Hospital Clinic, Barcelona, Spain
| | - Peter R. Schofield
- Neuroscience Research Australia, Sydney, New South Wales, Australia; School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Allan Levey
- Department of Neurology, Emory University, Atlanta, GA, USA
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany. Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Colin L. Masters
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia; University of Melbourne, Melbourne, Victoria, Australia
| | - Anne M. Fagan
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA; Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Randall J. Bateman
- Dominantly Inherited Alzheimer Network, Washington University School of Medicine, St. Louis, MO, USA; Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ralph N. Martins
- Macquarie Medical School, Macquarie University, North Ryde, NSW 2019, Australia; School of Medical Sciences, Edith Cowan University, Sarich Neuroscience Research Institute, Nedlands, WA 6009, Australia; The Cooperative Research Centre for Mental Health, Carlton South, Australia; KaRa Institute of Neurological Disease, Sydney, Macquarie Park, Australia; Australian Alzheimer’s Research Foundation, Nedlands, WA, Australia
| | - Charlotte E. Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, programme Neurodegeneration, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, The Netherlands
| | | |
Collapse
|
33
|
Ferreiro AL, Choi J, Ryou J, Newcomer EP, Thompson R, Bollinger RM, Hall-Moore C, Ndao IM, Sax L, Benzinger TLS, Stark SL, Holtzman DM, Fagan AM, Schindler SE, Cruchaga C, Butt OH, Morris JC, Tarr PI, Ances BM, Dantas G. Gut microbiome composition may be an indicator of preclinical Alzheimer's disease. Sci Transl Med 2023; 15:eabo2984. [PMID: 37315112 PMCID: PMC10680783 DOI: 10.1126/scitranslmed.abo2984] [Citation(s) in RCA: 84] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 05/26/2023] [Indexed: 06/16/2023]
Abstract
Alzheimer's disease (AD) pathology is thought to progress from normal cognition through preclinical disease and ultimately to symptomatic AD with cognitive impairment. Recent work suggests that the gut microbiome of symptomatic patients with AD has an altered taxonomic composition compared with that of healthy, cognitively normal control individuals. However, knowledge about changes in the gut microbiome before the onset of symptomatic AD is limited. In this cross-sectional study that accounted for clinical covariates and dietary intake, we compared the taxonomic composition and gut microbial function in a cohort of 164 cognitively normal individuals, 49 of whom showed biomarker evidence of early preclinical AD. Gut microbial taxonomic profiles of individuals with preclinical AD were distinct from those of individuals without evidence of preclinical AD. The change in gut microbiome composition correlated with β-amyloid (Aβ) and tau pathological biomarkers but not with biomarkers of neurodegeneration, suggesting that the gut microbiome may change early in the disease process. We identified specific gut bacterial taxa associated with preclinical AD. Inclusion of these microbiome features improved the accuracy, sensitivity, and specificity of machine learning classifiers for predicting preclinical AD status when tested on a subset of the cohort (65 of the 164 participants). Gut microbiome correlates of preclinical AD neuropathology may improve our understanding of AD etiology and may help to identify gut-derived markers of AD risk.
Collapse
Affiliation(s)
- Aura L. Ferreiro
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
- Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - JooHee Choi
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jian Ryou
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Erin P. Newcomer
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Regina Thompson
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rebecca M. Bollinger
- Program in Occupational Therapy, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carla Hall-Moore
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - I. Malick Ndao
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Laurie Sax
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tammie L. S. Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Susan L. Stark
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Program in Occupational Therapy, Washington University School of Medicine, St. Louis, MO 63110, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David M. Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anne M. Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Suzanne E. Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carlos Cruchaga
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
- NeuroGenomics and Informatics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Omar H. Butt
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Phillip I. Tarr
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Beau M. Ances
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Gautam Dantas
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
- Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
34
|
Horie K, Li Y, Barthélemy NR, Gordon BA, Hassenstab J, Benzinger TL, Fagan AM, Morris JC, Karch CM, Xiong C, Allegri R, Mendez PC, Ikeuchi T, Kasuga K, Noble J, Farlow M, Chhatwal J, Day GS, Schofield PR, Masters CL, Levin J, Jucker M, Lee JH, Hoon Roh J, Sato C, Sachdev P, Koyama A, Reyderman L, Bateman RJ, McDade E. Change in Cerebrospinal Fluid Tau Microtubule Binding Region Detects Symptom Onset, Cognitive Decline, Tangles, and Atrophy in Dominantly Inherited Alzheimer's Disease. Ann Neurol 2023; 93:1158-1172. [PMID: 36843330 PMCID: PMC10238659 DOI: 10.1002/ana.26620] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/28/2023]
Abstract
OBJECTIVE Identifying cerebrospinal fluid measures of the microtubule binding region of tau (MTBR-tau) species that reflect tau aggregation could provide fluid biomarkers that track Alzheimer's disease related neurofibrillary tau pathological changes. We examined the cerebrospinal fluid (CSF) MTBR-tau species in dominantly inherited Alzheimer's disease (DIAD) mutation carriers to assess the association with Alzheimer's disease (AD) biomarkers and clinical symptoms. METHODS Cross-sectional and longitudinal CSF from 229 DIAD mutation carriers and 130 mutation non-carriers had sequential characterization of N-terminal/mid-domain phosphorylated tau (p-tau) followed by MTBR-tau species and tau positron emission tomography (tau PET), other soluble tau and amyloid biomarkers, comprehensive clinical and cognitive assessments, and brain magnetic resonance imaging of atrophy. RESULTS CSF MTBR-tau species located within the putative "border" region and one species corresponding to the "core" region of aggregates in neurofibrillary tangles (NFTs) increased during the presymptomatic stage and decreased during the symptomatic stage. The "border" MTBR-tau species were associated with amyloid pathology and CSF p-tau; whereas the "core" MTBR-tau species were associated stronger with tau PET and CSF measures of neurodegeneration. The ratio of the border to the core species provided a continuous measure of increasing amounts that tracked clinical progression and NFTs. INTERPRETATION Changes in CSF soluble MTBR-tau species preceded the onset of dementia, tau tangle increase, and atrophy in DIAD. The ratio of 4R-specific MTBR-tau (border) to the NFT (core) MTBR-tau species corresponds to the pathology of NFTs in DIAD and change with disease progression. The dynamics between different MTBR-tau species in the CSF may serve as a marker of tau-related disease progression and target engagement of anti-tau therapeutics. ANN NEUROL 2023;93:1158-1172.
Collapse
Affiliation(s)
- Kanta Horie
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Eisai Inc., Nutley, NJ, 07110, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Nicolas R. Barthélemy
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Brian A. Gordon
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Tammie. L.S. Benzinger
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Anne M. Fagan
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Celeste M. Karch
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Ricardo Allegri
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI) Instituto de Investigaciones Neurológicas Raúl Correa, Buenos Aires, Argentina
| | - Patricio Chrem Mendez
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI) Instituto de Investigaciones Neurológicas Raúl Correa, Buenos Aires, Argentina
| | | | | | - James Noble
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, G.H. Sergievsky Center, Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032 USA
| | - Martin Farlow
- Department of Neurology, Indiana University, Indianapolis, IN 46202, USA
| | - Jasmeer Chhatwal
- Massachusetts General Hospital, Harvard Medical School Boston, MA 02114, USA
| | - Gregory S. Day
- Department of Neurology, Mayo Clinic in Florida, Jacksonville, FL 32224, USA
| | - Peter R. Schofield
- Neuroscience Research Australia, Sydney, 2031 NSW, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, 2052 NSW, Australia
| | - Colin L. Masters
- The Florey Institute and the University of Melbourne, Parkville, Victoria 3010, Australia
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE) Munich, Marchioninistr 15, D-83177 Munchen, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurology, Ludwig-Maximilians Universität München, Marchioninistr 15, 83177 Munich, Germany
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases (DZNE) Tübingen; and Hertie-Institute for Clinical Brain Research, University of Tübingen, D-72076 Tübingen, Germany
| | - Jae-Hong Lee
- Department of Neurology, Asan Medical Center, Seoul 05505, Korea
| | - Jee Hoon Roh
- Departments of Biomedical Sciences, Physiology, and Neurology, Korea University College of Medicine, Seoul 02841, Korea
| | - Chihiro Sato
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | | | | | | | - Randall J. Bateman
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Eric McDade
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | | |
Collapse
|
35
|
Sanaat A, Shooli H, Böhringer AS, Sadeghi M, Shiri I, Salimi Y, Ginovart N, Garibotto V, Arabi H, Zaidi H. A cycle-consistent adversarial network for brain PET partial volume correction without prior anatomical information. Eur J Nucl Med Mol Imaging 2023; 50:1881-1896. [PMID: 36808000 PMCID: PMC10199868 DOI: 10.1007/s00259-023-06152-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 02/12/2023] [Indexed: 02/23/2023]
Abstract
PURPOSE Partial volume effect (PVE) is a consequence of the limited spatial resolution of PET scanners. PVE can cause the intensity values of a particular voxel to be underestimated or overestimated due to the effect of surrounding tracer uptake. We propose a novel partial volume correction (PVC) technique to overcome the adverse effects of PVE on PET images. METHODS Two hundred and twelve clinical brain PET scans, including 50 18F-Fluorodeoxyglucose (18F-FDG), 50 18F-Flortaucipir, 36 18F-Flutemetamol, and 76 18F-FluoroDOPA, and their corresponding T1-weighted MR images were enrolled in this study. The Iterative Yang technique was used for PVC as a reference or surrogate of the ground truth for evaluation. A cycle-consistent adversarial network (CycleGAN) was trained to directly map non-PVC PET images to PVC PET images. Quantitative analysis using various metrics, including structural similarity index (SSIM), root mean squared error (RMSE), and peak signal-to-noise ratio (PSNR), was performed. Furthermore, voxel-wise and region-wise-based correlations of activity concentration between the predicted and reference images were evaluated through joint histogram and Bland and Altman analysis. In addition, radiomic analysis was performed by calculating 20 radiomic features within 83 brain regions. Finally, a voxel-wise two-sample t-test was used to compare the predicted PVC PET images with reference PVC images for each radiotracer. RESULTS The Bland and Altman analysis showed the largest and smallest variance for 18F-FDG (95% CI: - 0.29, + 0.33 SUV, mean = 0.02 SUV) and 18F-Flutemetamol (95% CI: - 0.26, + 0.24 SUV, mean = - 0.01 SUV), respectively. The PSNR was lowest (29.64 ± 1.13 dB) for 18F-FDG and highest (36.01 ± 3.26 dB) for 18F-Flutemetamol. The smallest and largest SSIM were achieved for 18F-FDG (0.93 ± 0.01) and 18F-Flutemetamol (0.97 ± 0.01), respectively. The average relative error for the kurtosis radiomic feature was 3.32%, 9.39%, 4.17%, and 4.55%, while it was 4.74%, 8.80%, 7.27%, and 6.81% for NGLDM_contrast feature for 18F-Flutemetamol, 18F-FluoroDOPA, 18F-FDG, and 18F-Flortaucipir, respectively. CONCLUSION An end-to-end CycleGAN PVC method was developed and evaluated. Our model generates PVC images from the original non-PVC PET images without requiring additional anatomical information, such as MRI or CT. Our model eliminates the need for accurate registration or segmentation or PET scanner system response characterization. In addition, no assumptions regarding anatomical structure size, homogeneity, boundary, or background level are required.
Collapse
Affiliation(s)
- Amirhossein Sanaat
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva, Switzerland
| | - Hossein Shooli
- Persian Gulf Nuclear Medicine Research Center, Department of Molecular Imaging and Radionuclide Therapy (MIRT), Bushehr Medical University Hospital, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Andrew Stephen Böhringer
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva, Switzerland
| | - Maryam Sadeghi
- Department of Medical Statistics, Informatics and Health Economics, Medical University of Innsbruck, Schoepfstr. 41, Innsbruck, Austria
| | - Isaac Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva, Switzerland
| | - Yazdan Salimi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva, Switzerland
| | - Nathalie Ginovart
- Geneva University Neurocenter, University of Geneva, Geneva, Switzerland
- Department of Psychiatry, Geneva University, Geneva, Switzerland
- Department of Basic Neuroscience, Geneva University, Geneva, Switzerland
| | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva, Switzerland
- Geneva University Neurocenter, University of Geneva, Geneva, Switzerland
| | - Hossein Arabi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva, Switzerland
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva, Switzerland.
- Geneva University Neurocenter, University of Geneva, Geneva, Switzerland.
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, Groningen, Netherlands.
- Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
36
|
Liu G, Shen C, Qiu A. Amyloid-β Accumulation in Relation to Functional Connectivity in Aging: a Longitudinal Study. Neuroimage 2023; 275:120146. [PMID: 37127190 DOI: 10.1016/j.neuroimage.2023.120146] [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: 01/15/2023] [Revised: 04/11/2023] [Accepted: 04/28/2023] [Indexed: 05/03/2023] Open
Abstract
The brain undergoes many changes at pathological and functional levels in healthy aging. This study employed a longitudinal and multimodal imaging dataset from the OASIS-3 study (n=300) and explored possible relationships between amyloid beta (Aβ) accumulation and functional brain organization over time in healthy aging. We used positron emission tomography (PET) with Pittsburgh compound-B (PIB) to quantify the Aβ accumulation in the brain and resting-state functional MRI (rs-fMRI) to measure functional connectivity (FC) among brain regions. Each participant had at least 2 to 3 follow-up visits. A linear mixed-effect model was used to examine longitudinal changes of Aβ accumulation and FC throughout the whole brain. We found that the limbic and frontoparietal networks had a greater annual Aβ accumulation and a slower decline in FC in aging. Additionally, the amount of the Aβ deposition in the amygdala network at baseline slowed down the decline in its FC in aging. Furthermore, the functional connectivity of the limbic, default mode network (DMN), and frontoparietal networks accelerated the Aβ propagation across their functionally highly connected regions. The functional connectivity of the somatomotor and visual networks accelerated the Aβ propagation across the brain regions in the limbic, frontoparietal, and DMN networks. These findings suggested that the slower decline in the functional connectivity of the functional hubs may compensate for their greater Aβ accumulation in aging. The Aβ propagation from one brain region to the other may depend on their functional connectivity strength.
Collapse
Affiliation(s)
- Guodong Liu
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | - Chenye Shen
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | - Anqi Qiu
- Department of Biomedical Engineering, National University of Singapore, Singapore; NUS (Suzhou) Research Institute, National University of Singapore, China; The N.1 Institute for Health, National University of Singapore, Singapore; Institute of Data Science, National University of Singapore, Singapore; Department of Biomedical Engineering, the Johns Hopkins University, USA.
| |
Collapse
|
37
|
Barthélemy NR, Saef B, Li Y, Gordon BA, He Y, Horie K, Stomrud E, Salvadó G, Janelidze S, Sato C, Ovod V, Henson RL, Fagan AM, Benzinger TLS, Xiong C, Morris JC, Hansson O, Bateman RJ, Schindler SE. CSF tau phosphorylation occupancies at T217 and T205 represent improved biomarkers of amyloid and tau pathology in Alzheimer's disease. NATURE AGING 2023; 3:391-401. [PMID: 37117788 PMCID: PMC10154225 DOI: 10.1038/s43587-023-00380-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 02/03/2023] [Indexed: 04/30/2023]
Abstract
Cerebrospinal fluid (CSF) amyloid-β peptide (Aβ)42/Aβ40 and the concentration of tau phosphorylated at site 181 (p-tau181) are well-established biomarkers of Alzheimer's disease (AD). The present study used mass spectrometry to measure concentrations of nine phosphorylated and five nonphosphorylated tau species and phosphorylation occupancies (percentage phosphorylated/nonphosphorylated) at ten sites. In the present study we show that, in 750 individuals with a median age of 71.2 years, CSF pT217/T217 predicted the presence of brain amyloid by positron emission tomography (PET) slightly better than Aβ42/Aβ40 (P = 0.02). Furthermore, for individuals with positive brain amyloid by PET (n = 263), CSF pT217/T217 was more strongly correlated with the amount of amyloid (Spearman's ρ = 0.69) than Aβ42/Aβ40 (ρ = -0.42, P < 0.0001). In two independent cohorts of participants with symptoms of AD dementia (n = 55 and n = 90), CSF pT217/T217 and pT205/T205 were better correlated with tau PET measures than CSF p-tau181 concentration. These findings suggest that CSF pT217/T217 and pT205/T205 represent improved CSF biomarkers of amyloid and tau pathology in AD.
Collapse
Affiliation(s)
- Nicolas R Barthélemy
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA.
| | - Benjamin Saef
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yingxin He
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Kanta Horie
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Chihiro Sato
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Vitaliy Ovod
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Rachel L Henson
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne M Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Chengjie Xiong
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
38
|
Zhukovsky P, Coughlan G, Buckley R, Grady C, Voineskos AN. Connectivity between default mode and frontoparietal networks mediates the association between global amyloid-β and episodic memory. Hum Brain Mapp 2023; 44:1147-1157. [PMID: 36420978 PMCID: PMC9875925 DOI: 10.1002/hbm.26148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/20/2022] [Accepted: 10/28/2022] [Indexed: 11/25/2022] Open
Abstract
Βeta-amyloid (Aβ) is a neurotoxic protein that deposits early in the pathogenesis of preclinical Alzheimer's disease. We aimed to identify network connectivity that may alter the negative effect of Aβ on cognition. Following assessment of memory performance, resting-state fMRI, and mean cortical PET-Aβ, a total of 364 older adults (286 with clinical dementia rating [CDR-0], 59 with CDR-0.5 and 19 with CDR-1, mean age: 74.0 ± 6.4 years) from the OASIS-3 sample were included in the analysis. Across all participants, a partial least squares regression showed that lower connectivity between posterior medial default mode and frontoparietal networks, higher within-default mode, and higher visual-motor connectivity predict better episodic memory. These connectivities partially mediate the effect of Aβ on episodic memory. These results suggest that connectivity strength between the precuneus cortex and the superior frontal gyri may alter the negative effect of Aβ on episodic memory. In contrast, education was associated with different functional connectivity patterns. In conclusion, functional characteristics of specific brain networks may help identify amyloid-positive individuals with a higher likelihood of memory decline, with implications for AD clinical trials.
Collapse
Affiliation(s)
- Peter Zhukovsky
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, Toronto, Canada
| | - Gillian Coughlan
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rachel Buckley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Melbourne School of Psychological Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Cheryl Grady
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Ontario, Canada
| | - Aristotle N Voineskos
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, Toronto, Canada
| |
Collapse
|
39
|
Goyal MS, Blazey T, Metcalf NV, McAvoy MP, Strain JF, Rahmani M, Durbin TJ, Xiong C, Benzinger TLS, Morris JC, Raichle ME, Vlassenko AG. Brain aerobic glycolysis and resilience in Alzheimer disease. Proc Natl Acad Sci U S A 2023; 120:e2212256120. [PMID: 36745794 PMCID: PMC9963219 DOI: 10.1073/pnas.2212256120] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 01/04/2023] [Indexed: 02/08/2023] Open
Abstract
The distribution of brain aerobic glycolysis (AG) in normal young adults correlates spatially with amyloid-beta (Aβ) deposition in individuals with symptomatic and preclinical Alzheimer disease (AD). Brain AG decreases with age, but the functional significance of this decrease with regard to the development of AD symptomatology is poorly understood. Using PET measurements of regional blood flow, oxygen consumption, and glucose utilization-from which we derive AG-we find that cognitive impairment is strongly associated with loss of the typical youthful pattern of AG. In contrast, amyloid positivity without cognitive impairment was associated with preservation of youthful brain AG, which was even higher than that seen in cognitively unimpaired, amyloid negative adults. Similar findings were not seen for blood flow nor oxygen consumption. Finally, in cognitively unimpaired adults, white matter hyperintensity burden was found to be specifically associated with decreased youthful brain AG. Our results suggest that AG may have a role in the resilience and/or response to early stages of amyloid pathology and that age-related white matter disease may impair this process.
Collapse
Affiliation(s)
- Manu S. Goyal
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO63110
- Department of Neurology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO63108
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO63110
| | - Tyler Blazey
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
| | - Nicholas V. Metcalf
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
| | - Mark P. McAvoy
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
- Program in Occupational Therapy, Washington University School of Medicine, St. Louis, MO63108
| | - Jeremy F. Strain
- Department of Neurology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
| | - Maryam Rahmani
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
| | - Tony J. Durbin
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
| | - Chengjie Xiong
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO63108
| | - Tammie L.-S. Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO63108
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO63110
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO63108
| | - Marcus E. Raichle
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO63110
- Department of Neurology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO63108
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO63110
- Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO63130
- Department of Psychology & Brain Science, Washington University School of Medicine, St. Louis, MO63130
| | - Andrei G. Vlassenko
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO63108
| |
Collapse
|
40
|
Data-Driven Phenotyping of Alzheimer's Disease under Epigenetic Conditions Using Partial Volume Correction of PET Studies and Manifold Learning. Biomedicines 2023; 11:biomedicines11020273. [PMID: 36830810 PMCID: PMC9953610 DOI: 10.3390/biomedicines11020273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/10/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. An increasing number of studies have confirmed epigenetic changes in AD. Consequently, a robust phenotyping mechanism must take into consideration the environmental effects on the patient in the generation of phenotypes. Positron Emission Tomography (PET) is employed for the quantification of pathological amyloid deposition in brain tissues. The objective is to develop a new methodology for the hyperparametric analysis of changes in cognitive scores and PET features to test for there being multiple AD phenotypes. We used a computational method to identify phenotypes in a retrospective cohort study (532 subjects), using PET and Magnetic Resonance Imaging (MRI) images and neuropsychological assessments, to develop a novel computational phenotyping method that uses Partial Volume Correction (PVC) and subsets of neuropsychological assessments in a non-biased fashion. Our pipeline is based on a Regional Spread Function (RSF) method for PVC and a t-distributed Stochastic Neighbor Embedding (t-SNE) manifold. The results presented demonstrate that (1) the approach to data-driven phenotyping is valid, (2) the different techniques involved in the pipelines produce different results, and (3) they permit us to identify the best phenotyping pipeline. The method identifies three phenotypes and permits us to analyze them under epigenetic conditions.
Collapse
|
41
|
Chen CD, Ponisio MR, Lang JA, Flores S, Schindler SE, Fagan AM, Morris JC, Benzinger TL. Comparing Tau PET Visual Interpretation with Tau PET Quantification, Cerebrospinal Fluid Biomarkers, and Longitudinal Clinical Assessment. J Alzheimers Dis 2023; 93:765-777. [PMID: 37092225 PMCID: PMC10200228 DOI: 10.3233/jad-230032] [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] [Accepted: 03/14/2023] [Indexed: 04/25/2023]
Abstract
BACKGROUND 18F-flortaucipir PET received FDA approval to visualize aggregated neurofibrillary tangles (NFTs) in brains of adult patients with cognitive impairment being evaluated for Alzheimer's disease (AD). However, manufacturer's guidelines for visual interpretation of 18F-flortaucipir PET differ from how 18F-flortaucipir PET has been measured in research settings using standardized uptake value ratios (SUVRs). How visual interpretation relates to 18F-flortaucipir PET SUVR, cerebrospinal fluid (CSF) biomarkers, or longitudinal clinical assessment is not well understood. OBJECTIVE We compare various diagnostic methods in participants enrolled in longitudinal observational studies of aging and memory (n = 189, 23 were cognitively impaired). METHODS Participants had tau PET, Aβ PET, MRI, and clinical and cognitive evaluation within 18 months (n = 189); the majority (n = 144) also underwent lumbar puncture. Two radiologists followed manufacturer's guidelines for 18F-flortaucipir PET visual interpretation. RESULTS Visual interpretation had high agreement with SUVR (98.4%)and moderate agreement with CSF p-tau181 (86.1%). Two participants demonstrated 18F-flortaucipir uptake from meningiomas. Visual interpretation could not predict follow-up clinical assessment in 9.52% of cases. CONCLUSION Visual interpretation was highly consistent with SUVR (discordant participants had hemorrhagic infarcts or occipital-predominant AD NFT deposition) and moderately consistent with CSF p-tau181 (discordant participants had AD pathophysiology not detectable on tau PET). However, close association between AD NFT deposition and clinical onset in group-level studies does not necessarily hold at the individual level, with discrepancies arising from atypical AD, vascular dementia, or frontotemporal dementia. A better understanding of relationships across imaging, CSF biomarkers, and clinical assessment is needed to provide appropriate diagnoses for these individuals.
Collapse
Affiliation(s)
- Charles D. Chen
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Maria Rosana Ponisio
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Jordan A. Lang
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Shaney Flores
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Anne M. Fagan
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - John C. Morris
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Tammie L.S. Benzinger
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
42
|
Petersen KK, Ezzati A, Lipton RB, Gordon BA, Hassenstab J, Morris JC, Grober E. Associations of Stages of Objective Memory Impairment with Cerebrospinal Fluid and Neuroimaging Biomarkers of Alzheimer's Disease. J Prev Alzheimers Dis 2023; 10:112-119. [PMID: 36641615 PMCID: PMC9841119 DOI: 10.14283/jpad.2022.98] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVE To investigate cerebrospinal fluid (CSF) and neuroimaging correlates of Stages of Objective Memory Impairment (SOMI) based on Free and Cued Selective Reminding Test (FCSRT) performance, and to evaluate the effect of APOE ε4 status on this relationship. METHODS Data from 586 cognitively unimpaired individuals who had FCSRT, CSF, and volumetric magnetic resonance imaging (MRI) measures available was used. We compared CSF measures of β-amyloid (Aβ42/Aβ40 ratio), phosphorylated tau (p-Tau181), total tau (t-Tau), hippocampal volume, and PIB-PET mean cortical binding potential with partial volume correction (MCBP) among SOMI groups in the whole sample and in subsamples stratified by APOE ε4 status. RESULTS Participants had a mean age of 67.4 (SD=9.1) years, had 16.1 (SD=2.6) years of education, 57.0% were female, and 33.8% were APOE ε4 positive. In the entire sample, there was no significant difference between SOMI stages in Aβ42/Aβ40 ratio, p-Tau181, t-Tau, or PIB-PET MCBP when adjusted for age, sex, and education. However, higher SOMI stages had smaller hippocampal volume (F=3.29, p=0.020). In the stratified sample based on APOE ε4 status, in APOE ε4 positive individuals, higher SOMI stages had higher p-Tau181 (F=2.94, p=0.034) higher t-Tau (F=3.41, p=0.019), and smaller hippocampal volume (F=5.78, p<0.001). There were no significant differences in CSF or imaging biomarkers between SOMI groups in the APOE ε4 negative subsample. CONCLUSION Cognitively normal older individuals with higher SOMI stages have higher in-vivo tau and neurodegenerative pathology only in APOE ε4 carriers. These original results indicate the potential usefulness of the SOMI staging system in assessing of tau and neurodegenerative pathology.
Collapse
Affiliation(s)
- K K Petersen
- Kellen K. Petersen, Albert Einstein College of Medicine, 1225 Morris Park Avenue, Bronx, NY 10461, USA,
| | | | | | | | | | | | | |
Collapse
|
43
|
Wisch JK, Gordon BA, Boerwinkle AH, Luckett PH, Bollinger JG, Ovod V, Li Y, Henson RL, West T, Meyer MR, Kirmess KM, Benzinger TL, Fagan AM, Morris JC, Bateman RJ, Ances BM, Schindler SE. Predicting continuous amyloid PET values with CSF and plasma Aβ42/Aβ40. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12405. [PMID: 36874595 PMCID: PMC9980305 DOI: 10.1002/dad2.12405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/14/2022] [Accepted: 01/19/2023] [Indexed: 03/06/2023]
Abstract
Introduction Continuous measures of amyloid burden as measured by positron emission tomography (PET) are being used increasingly to stage Alzheimer's disease (AD). This study examined whether cerebrospinal fluid (CSF) and plasma amyloid beta (Aβ)42/Aβ40 could predict continuous values for amyloid PET. Methods CSF Aβ42 and Aβ40 were measured with automated immunoassays. Plasma Aβ42 and Aβ40 were measured with an immunoprecipitation-mass spectrometry assay. Amyloid PET was performed with Pittsburgh compound B (PiB). The continuous relationships of CSF and plasma Aβ42/Aβ40 with amyloid PET burden were modeled. Results Most participants were cognitively normal (427 of 491 [87%]) and the mean age was 69.0 ± 8.8 years. CSF Aβ42/Aβ40 predicted amyloid PET burden until a relatively high level of amyloid accumulation (69.8 Centiloids), whereas plasma Aβ42/Aβ40 predicted amyloid PET burden until a lower level (33.4 Centiloids). Discussion CSF Aβ42/Aβ40 predicts the continuous level of amyloid plaque burden over a wider range than plasma Aβ42/Aβ40 and may be useful in AD staging. Highlights Cerebrospinal fluid (CSF) amyloid beta (Aβ)42/Aβ40 predicts continuous amyloid positron emission tomography (PET) values up to a relatively high burden.Plasma Aβ42/Aβ40 is a comparatively dichotomous measure of brain amyloidosis.Models can predict regional amyloid PET burden based on CSF Aβ42/Aβ40.CSF Aβ42/Aβ40 may be useful in staging AD.
Collapse
Affiliation(s)
- Julie K. Wisch
- Department of NeurologyWashington University in Saint LouisSt. LouisMissouriUSA
| | - Brian A. Gordon
- Department of RadiologyWashington University in Saint LouisSt. LouisMissouriUSA
- Hope CenterWashington University in Saint LouisSt. LouisMissouriUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt LouisMissouriUSA
| | - Anna H. Boerwinkle
- Department of NeurologyWashington University in Saint LouisSt. LouisMissouriUSA
| | - Patrick H. Luckett
- Department of NeurologyWashington University in Saint LouisSt. LouisMissouriUSA
| | - James G. Bollinger
- Department of NeurologyWashington University in Saint LouisSt. LouisMissouriUSA
- The Tracy Family SILQ Center for Neurodegenerative BiologySt. LouisMissouriUSA
| | - Vitaliy Ovod
- Department of NeurologyWashington University in Saint LouisSt. LouisMissouriUSA
- The Tracy Family SILQ Center for Neurodegenerative BiologySt. LouisMissouriUSA
| | - Yan Li
- Department of RadiologyWashington University in Saint LouisSt. LouisMissouriUSA
| | - Rachel L. Henson
- Department of NeurologyWashington University in Saint LouisSt. LouisMissouriUSA
| | - Tim West
- C2N DiagnosticsSt. LouisMissouriUSA
| | | | | | - Tammie L.S. Benzinger
- Department of RadiologyWashington University in Saint LouisSt. LouisMissouriUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt LouisMissouriUSA
| | - Anne M. Fagan
- Department of NeurologyWashington University in Saint LouisSt. LouisMissouriUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt LouisMissouriUSA
| | - John C. Morris
- Department of NeurologyWashington University in Saint LouisSt. LouisMissouriUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt LouisMissouriUSA
| | - Randall J. Bateman
- Department of NeurologyWashington University in Saint LouisSt. LouisMissouriUSA
- The Tracy Family SILQ Center for Neurodegenerative BiologySt. LouisMissouriUSA
| | - Beau M. Ances
- Department of NeurologyWashington University in Saint LouisSt. LouisMissouriUSA
- Department of RadiologyWashington University in Saint LouisSt. LouisMissouriUSA
- Hope CenterWashington University in Saint LouisSt. LouisMissouriUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt LouisMissouriUSA
| | - Suzanne E. Schindler
- Department of NeurologyWashington University in Saint LouisSt. LouisMissouriUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt LouisMissouriUSA
| |
Collapse
|
44
|
Luckett PH, Chen C, Gordon BA, Wisch J, Berman SB, Chhatwal JP, Cruchaga C, Fagan AM, Farlow MR, Fox NC, Jucker M, Levin J, Masters CL, Mori H, Noble JM, Salloway S, Schofield PR, Brickman AM, Brooks WS, Cash DM, Fulham MJ, Ghetti B, Jack CR, Vöglein J, Klunk WE, Koeppe R, Su Y, Weiner M, Wang Q, Marcus D, Koudelis D, Mathurin NJ, Cash L, Hornbeck R, Xiong C, Perrin RJ, Karch CM, Hassenstab J, McDade E, Morris JC, Benzinger TL, Bateman RJ, Ances BM. Biomarker clustering in autosomal dominant Alzheimer's disease. Alzheimers Dement 2023; 19:274-284. [PMID: 35362200 PMCID: PMC9525451 DOI: 10.1002/alz.12661] [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: 11/02/2021] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 01/18/2023]
Abstract
INTRODUCTION As the number of biomarkers used to study Alzheimer's disease (AD) continues to increase, it is important to understand the utility of any given biomarker, as well as what additional information a biomarker provides when compared to others. METHODS We used hierarchical clustering to group 19 cross-sectional biomarkers in autosomal dominant AD. Feature selection identified biomarkers that were the strongest predictors of mutation status and estimated years from symptom onset (EYO). Biomarkers identified included clinical assessments, neuroimaging, cerebrospinal fluid amyloid, and tau, and emerging biomarkers of neuronal integrity and inflammation. RESULTS Three primary clusters were identified: neurodegeneration, amyloid/tau, and emerging biomarkers. Feature selection identified amyloid and tau measures as the primary predictors of mutation status and EYO. Emerging biomarkers of neuronal integrity and inflammation were relatively weak predictors. DISCUSSION These results provide novel insight into our understanding of the relationships among biomarkers and the staging of biomarkers based on disease progression.
Collapse
Affiliation(s)
| | - Charlie Chen
- Washington University in St. Louis, St. Louis, Missouri, USA
| | - Brian A. Gordon
- Washington University in St. Louis, St. Louis, Missouri, USA
| | - Julie Wisch
- Washington University in St. Louis, St. Louis, Missouri, USA
| | | | - Jasmeer P. Chhatwal
- Brigham and Women’s Hospital, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Carlos Cruchaga
- Washington University in St. Louis, St. Louis, Missouri, USA
| | - Anne M. Fagan
- Washington University in St. Louis, St. Louis, Missouri, USA
| | | | - Nick C. Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Mathias Jucker
- German Center for Neurodegenerative Disease, Tübingen, Germany
- Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Colin L. Masters
- Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Hiroshi Mori
- Osaka City University Medical School, Nagaoka Sutoku University, Abenoku, Osaka, Japan
| | - James M. Noble
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, G.H. Sergievsky Center, and Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Stephen Salloway
- Butler Hospital and Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Peter R. Schofield
- Neuroscience Research Australia, Randwick, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Adam M. Brickman
- Department of Molecular Imaging, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- The University of Sydney, Sydney, New South Wales, Australia
| | - William S. Brooks
- Neuroscience Research Australia, Randwick, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - David M. Cash
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Michael J. Fulham
- Department of Molecular Imaging, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- The University of Sydney, Sydney, New South Wales, Australia
| | | | | | - Jonathan Vöglein
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | | | | | - Yi Su
- Banner Alzheimer Institute, Phoenix, Arizona, USA
| | - Michael Weiner
- University of California San Francisco, San Francisco, California, USA
- San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - Qing Wang
- Washington University in St. Louis, St. Louis, Missouri, USA
| | - Daniel Marcus
- Washington University in St. Louis, St. Louis, Missouri, USA
| | | | | | - Lisa Cash
- Washington University in St. Louis, St. Louis, Missouri, USA
| | - Russ Hornbeck
- Washington University in St. Louis, St. Louis, Missouri, USA
| | - Chengjie Xiong
- Washington University in St. Louis, St. Louis, Missouri, USA
| | | | | | | | - Eric McDade
- Washington University in St. Louis, St. Louis, Missouri, USA
| | - John C. Morris
- Washington University in St. Louis, St. Louis, Missouri, USA
| | | | | | - Beau M. Ances
- Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
45
|
Alosco ML, Su Y, Stein TD, Protas H, Cherry JD, Adler CH, Balcer LJ, Bernick C, Pulukuri SV, Abdolmohammadi B, Coleman MJ, Palmisano JN, Tripodis Y, Mez J, Rabinovici GD, Marek KL, Beach TG, Johnson KA, Huber BR, Koerte I, Lin AP, Bouix S, Cummings JL, Shenton ME, Reiman EM, McKee AC, Stern RA. Associations between near end-of-life flortaucipir PET and postmortem CTE-related tau neuropathology in six former American football players. Eur J Nucl Med Mol Imaging 2023; 50:435-452. [PMID: 36152064 PMCID: PMC9816291 DOI: 10.1007/s00259-022-05963-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/01/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE Flourine-18-flortaucipir tau positron emission tomography (PET) was developed for the detection for Alzheimer's disease. Human imaging studies have begun to investigate its use in chronic traumatic encephalopathy (CTE). Flortaucipir-PET to autopsy correlation studies in CTE are needed for diagnostic validation. We examined the association between end-of-life flortaucipir PET and postmortem neuropathological measurements of CTE-related tau in six former American football players. METHODS Three former National Football League players and three former college football players who were part of the DIAGNOSE CTE Research Project died and agreed to have their brains donated. The six players had flortaucipir (tau) and florbetapir (amyloid) PET prior to death. All brains from the deceased participants were neuropathologically evaluated for the presence of CTE. On average, the participants were 59.0 (SD = 9.32) years of age at time of PET. PET scans were acquired 20.33 (SD = 13.08) months before their death. Using Spearman correlation analyses, we compared flortaucipir standard uptake value ratios (SUVRs) to digital slide-based AT8 phosphorylated tau (p-tau) density in a priori selected composite cortical, composite limbic, and thalamic regions-of-interest (ROIs). RESULTS Four brain donors had autopsy-confirmed CTE, all with high stage disease (n = 3 stage III, n = 1 stage IV). Three of these four met criteria for the clinical syndrome of CTE, known as traumatic encephalopathy syndrome (TES). Two did not have CTE at autopsy and one of these met criteria for TES. Concomitant pathology was only present in one of the non-CTE cases (Lewy body) and one of the CTE cases (motor neuron disease). There was a strong association between flortaucipir SUVRs and p-tau density in the composite cortical (ρ = 0.71) and limbic (ρ = 0.77) ROIs. Although there was a strong association in the thalamic ROI (ρ = 0.83), this is a region with known off-target binding. SUVRs were modest and CTE and non-CTE cases had overlapping SUVRs and discordant p-tau density for some regions. CONCLUSIONS Flortaucipir-PET could be useful for detecting high stage CTE neuropathology, but specificity to CTE p-tau is uncertain. Off-target flortaucipir binding in the hippocampus and thalamus complicates interpretation of these associations. In vivo biomarkers that can detect the specific p-tau of CTE across the disease continuum are needed.
Collapse
Affiliation(s)
- Michael L Alosco
- Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Yi Su
- Banner Alzheimer's Institute, Arizona State University, and Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Thor D Stein
- Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
| | - Hillary Protas
- Banner Alzheimer's Institute, Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Jonathan D Cherry
- Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Laura J Balcer
- Departments of Neurology, Population Health and Ophthalmology, NYU Grossman School of Medicine, New York, NY, USA
| | - Charles Bernick
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Surya Vamsi Pulukuri
- Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Bobak Abdolmohammadi
- Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Michael J Coleman
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, USA
| | - Joseph N Palmisano
- Biostatistics and Epidemiology Data Analytics Center (BEDAC), Boston University School of Public Health, Boston, MA, USA
| | - Yorghos Tripodis
- Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Jesse Mez
- Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Gil D Rabinovici
- Memory & Aging Center, Departments of Neurology, Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Kenneth L Marek
- Institute for Neurodegenerative Disorders, Invicro, LLC, New Haven, CT, USA
| | - Thomas G Beach
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Keith A Johnson
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Boston, MA, USA
- Brigham and Women's Hospital, Boston, MA, USA
| | - Bertrand Russell Huber
- Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
- National Center for PTSD, VA Boston Healthcare, Jamaica Plain, MA, USA
| | - Inga Koerte
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Ludwig Maximilians University, Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig Maximilians University, Munich, Germany
- NICUM (NeuroImaging Core Unit Munich), Ludwig Maximilians University, Munich, Germany
| | - Alexander P Lin
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, USA
- Center for Clinical Spectroscopy, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sylvain Bouix
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, USA
| | - Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Martha E Shenton
- VA Boston Healthcare System, Boston, MA, USA
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, USA
- Brigham and Women's Hospital, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Eric M Reiman
- Banner Alzheimer's Institute, University of Arizona, Arizona State University, Translational Genomics Research Institute, and Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Ann C McKee
- Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
| | - Robert A Stern
- Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Department of Neurology, Boston University School of Medicine, Boston, MA, USA.
- Departments of Neurosurgery, and Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
46
|
Long JM, Coble DW, Xiong C, Schindler SE, Perrin RJ, Gordon BA, Benzinger TLS, Grant E, Fagan AM, Harari O, Cruchaga C, Holtzman DM, Morris JC. Preclinical Alzheimer's disease biomarkers accurately predict cognitive and neuropathological outcomes. Brain 2022; 145:4506-4518. [PMID: 35867858 PMCID: PMC10200309 DOI: 10.1093/brain/awac250] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/30/2022] [Accepted: 07/20/2022] [Indexed: 01/24/2023] Open
Abstract
Alzheimer's disease biomarkers are widely accepted as surrogate markers of underlying neuropathological changes. However, few studies have evaluated whether preclinical Alzheimer's disease biomarkers predict Alzheimer's neuropathology at autopsy. We sought to determine whether amyloid PET imaging or CSF biomarkers accurately predict cognitive outcomes and Alzheimer's disease neuropathological findings. This study included 720 participants, 42-91 years of age, who were enrolled in longitudinal studies of memory and aging in the Washington University Knight Alzheimer Disease Research Center and were cognitively normal at baseline, underwent amyloid PET imaging and/or CSF collection within 1 year of baseline clinical assessment, and had subsequent clinical follow-up. Cognitive status was assessed longitudinally by Clinical Dementia Rating®. Biomarker status was assessed using predefined cut-offs for amyloid PET imaging or CSF p-tau181/amyloid-β42. Subsequently, 57 participants died and underwent neuropathologic examination. Alzheimer's disease neuropathological changes were assessed using standard criteria. We assessed the predictive value of Alzheimer's disease biomarker status on progression to cognitive impairment and for presence of Alzheimer's disease neuropathological changes. Among cognitively normal participants with positive biomarkers, 34.4% developed cognitive impairment (Clinical Dementia Rating > 0) as compared to 8.4% of those with negative biomarkers. Cox proportional hazards modelling indicated that preclinical Alzheimer's disease biomarker status, APOE ɛ4 carrier status, polygenic risk score and centred age influenced risk of developing cognitive impairment. Among autopsied participants, 90.9% of biomarker-positive participants and 8.6% of biomarker-negative participants had Alzheimer's disease neuropathological changes. Sensitivity was 87.0%, specificity 94.1%, positive predictive value 90.9% and negative predictive value 91.4% for detection of Alzheimer's disease neuropathological changes by preclinical biomarkers. Single CSF and amyloid PET baseline biomarkers were also predictive of Alzheimer's disease neuropathological changes, as well as Thal phase and Braak stage of pathology at autopsy. Biomarker-negative participants who developed cognitive impairment were more likely to exhibit non-Alzheimer's disease pathology at autopsy. The detection of preclinical Alzheimer's disease biomarkers is strongly predictive of future cognitive impairment and accurately predicts presence of Alzheimer's disease neuropathology at autopsy.
Collapse
Affiliation(s)
- Justin M Long
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Dean W Coble
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Division of Biostatistics, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Chengjie Xiong
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Division of Biostatistics, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Suzanne E Schindler
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Richard J Perrin
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Brian A Gordon
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Tammie L S Benzinger
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Elizabeth Grant
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Division of Biostatistics, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Anne M Fagan
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Oscar Harari
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Carlos Cruchaga
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - David M Holtzman
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - John C Morris
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| |
Collapse
|
47
|
Jeong SH, Lee EC, Chung SJ, Lee HS, Jung JH, Sohn YH, Seong JK, Lee PH. Local striatal volume and motor reserve in drug-naïve Parkinson's disease. NPJ Parkinsons Dis 2022; 8:168. [PMID: 36470876 PMCID: PMC9722895 DOI: 10.1038/s41531-022-00429-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 11/11/2022] [Indexed: 12/12/2022] Open
Abstract
Motor reserve (MR) may explain why individuals with similar pathological changes show marked differences in motor deficits in Parkinson's disease (PD). In this study, we investigated whether estimated individual MR was linked to local striatal volume (LSV) in PD. We analyzed data obtained from 333 patients with drug naïve PD who underwent dopamine transporter scans and high-resolution 3-tesla T1-weighted structural magnetic resonance images. Using a residual model, we estimated individual MRs on the basis of initial UPDRS-III score and striatal dopamine depletion. We performed a correlation analysis between MR estimates and LSV. Furthermore, we assessed the effect of LSV, which is correlated with MR estimates, on the longitudinal increase in the levodopa-equivalent dose (LED) during the 4-year follow-up period using a linear mixed model. After controlling for intracranial volume, there was a significant positive correlation between LSV and MR estimates in the bilateral caudate, anterior putamen, and ventro-posterior putamen. The linear mixed model showed that the large local volume of anterior and ventro-posterior putamen was associated with the low requirement of LED initially and accelerated LED increment thereafter. The present study demonstrated that LSV is crucial to MR in early-stage PD, suggesting LSV as a neural correlate of MR in PD.
Collapse
Affiliation(s)
- Seong Ho Jeong
- grid.15444.300000 0004 0470 5454Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea ,grid.411627.70000 0004 0647 4151Department of Neurology, Inje University Sanggye Paik Hospital, Seoul, South Korea
| | - Eun-Chong Lee
- grid.222754.40000 0001 0840 2678School of Biomedical Engineering, Korea University, Seoul, South Korea
| | - Seok Jong Chung
- grid.15444.300000 0004 0470 5454Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea ,grid.413046.40000 0004 0439 4086Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, South Korea
| | - Hye Sun Lee
- grid.15444.300000 0004 0470 5454Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, South Korea
| | - Jin Ho Jung
- grid.411625.50000 0004 0647 1102Department of Neurology, Inje University Busan Paik Hospital, Seoul, South Korea
| | - Young H. Sohn
- grid.15444.300000 0004 0470 5454Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Joon-Kyung Seong
- grid.222754.40000 0001 0840 2678School of Biomedical Engineering, Korea University, Seoul, South Korea ,grid.222754.40000 0001 0840 2678Department of Artificial Intelligence, Korea University, Seoul, South Korea
| | - Phil Hyu Lee
- grid.15444.300000 0004 0470 5454Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea ,grid.15444.300000 0004 0470 5454Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
48
|
Shah J, Gao F, Li B, Ghisays V, Luo J, Chen Y, Lee W, Zhou Y, Benzinger TL, Reiman EM, Chen K, Su Y, Wu T. Deep residual inception encoder-decoder network for amyloid PET harmonization. Alzheimers Dement 2022; 18:2448-2457. [PMID: 35142053 PMCID: PMC9360199 DOI: 10.1002/alz.12564] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 12/02/2021] [Accepted: 12/05/2021] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Multiple positron emission tomography (PET) tracers are available for amyloid imaging, posing a significant challenge to consensus interpretation and quantitative analysis. We accordingly developed and validated a deep learning model as a harmonization strategy. METHOD A Residual Inception Encoder-Decoder Neural Network was developed to harmonize images between amyloid PET image pairs made with Pittsburgh Compound-B and florbetapir tracers. The model was trained using a dataset with 92 subjects with 10-fold cross validation and its generalizability was further examined using an independent external dataset of 46 subjects. RESULTS Significantly stronger between-tracer correlations (P < .001) were observed after harmonization for both global amyloid burden indices and voxel-wise measurements in the training cohort and the external testing cohort. DISCUSSION We proposed and validated a novel encoder-decoder based deep model to harmonize amyloid PET imaging data from different tracers. Further investigation is ongoing to improve the model and apply to additional tracers.
Collapse
Affiliation(s)
- Jay Shah
- ASU‐Mayo Center for Innovative ImagingArizona State University699 S. Mill Ave.TempeArizona85287USA
- School of Computing and Augmented IntelligenceArizona State University699 S. Mill Ave.TempeArizona85287USA
| | - Fei Gao
- ASU‐Mayo Center for Innovative ImagingArizona State University699 S. Mill Ave.TempeArizona85287USA
- School of Computing and Augmented IntelligenceArizona State University699 S. Mill Ave.TempeArizona85287USA
| | - Baoxin Li
- ASU‐Mayo Center for Innovative ImagingArizona State University699 S. Mill Ave.TempeArizona85287USA
- School of Computing and Augmented IntelligenceArizona State University699 S. Mill Ave.TempeArizona85287USA
| | - Valentina Ghisays
- Banner Alzheimer's Institute901 E. Willetta StreetPhoenixArizona85006USA
| | - Ji Luo
- Banner Alzheimer's Institute901 E. Willetta StreetPhoenixArizona85006USA
| | - Yinghua Chen
- Banner Alzheimer's Institute901 E. Willetta StreetPhoenixArizona85006USA
| | - Wendy Lee
- Banner Alzheimer's Institute901 E. Willetta StreetPhoenixArizona85006USA
| | - Yuxiang Zhou
- Department of RadiologyMayo Clinic at Arizona5777 E Mayo BlvdPhoenixArizona85054USA
| | - Tammie L.S. Benzinger
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St. Louis510 South Kingshighway BoulevardSt. LouisMissouri63110USA
| | - Eric M. Reiman
- Banner Alzheimer's Institute901 E. Willetta StreetPhoenixArizona85006USA
| | - Kewei Chen
- Banner Alzheimer's Institute901 E. Willetta StreetPhoenixArizona85006USA
| | - Yi Su
- ASU‐Mayo Center for Innovative ImagingArizona State University699 S. Mill Ave.TempeArizona85287USA
- School of Computing and Augmented IntelligenceArizona State University699 S. Mill Ave.TempeArizona85287USA
- Banner Alzheimer's Institute901 E. Willetta StreetPhoenixArizona85006USA
| | - Teresa Wu
- ASU‐Mayo Center for Innovative ImagingArizona State University699 S. Mill Ave.TempeArizona85287USA
- School of Computing and Augmented IntelligenceArizona State University699 S. Mill Ave.TempeArizona85287USA
| |
Collapse
|
49
|
Tarawneh R, Kasper RS, Sanford J, Phuah C, Hassenstab J, Cruchaga C. Vascular endothelial-cadherin as a marker of endothelial injury in preclinical Alzheimer disease. Ann Clin Transl Neurol 2022; 9:1926-1940. [PMID: 36342663 PMCID: PMC9735377 DOI: 10.1002/acn3.51685] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/02/2022] [Accepted: 10/10/2022] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVE Endothelial dysfunction is an early and prevalent pathology in Alzheimer disease (AD). We here investigate the value of vascular endothelial-cadherin (VEC) as a cerebrospinal fluid (CSF) marker of endothelial injury in preclinical AD. METHODS Cognitively normal participants (Clinical Dementia Rating [CDR] 0) from the Knight Washington University-ADRC were included in this study (n = 700). Preclinical Alzheimer's Cognitive Composite (PACC) scores, CSF VEC, tau, p-tau181, Aβ42/Aβ40, neurofilament light-chain (NFL) levels, and magnetic resonance imaging (MRI) assessments of white matter injury (WMI) were obtained from all participants. A subset of participants underwent brain amyloid imaging using positron emission tomography (amyloid-PET) (n = 534). Linear regression examined associations of CSF VEC with PACC and individual cognitive scores in preclinical AD. Mediation analyses examined whether CSF VEC mediated effects of CSF amyloid and tau markers on cognition in preclinical AD. RESULTS CSF VEC levels significantly correlated with PACC and individual cognitive scores in participants with amyloid (A+T±N±; n = 558) or those with amyloid and tau pathologies (A+T+N±; n = 259), after adjusting for covariates. CSF VEC also correlated with CSF measures of amyloid, tau, and neurodegeneration and global amyloid burden on amyloid-PET scans in our cohort. Importantly, our findings suggest that CSF VEC mediates associations of CSF Aβ42/Aβ40, p-tau181, and global amyloid burden with cognitive outcomes in preclinical AD. INTERPRETATION Our results support the utility of CSF VEC as a marker of endothelial injury in AD and highlight the importance of endothelial injury as an early pathology that contributes to cognitive impairment in even the earliest preclinical stages.
Collapse
Affiliation(s)
- Rawan Tarawneh
- Department of NeurologyUniversity of New MexicoAlbuquerqueNew MexicoUSA
- Center for Memory and AgingUniversity of New MexicoAlbuquerqueNew MexicoUSA
| | - Rachel S. Kasper
- Department of NeurologyUniversity of New MexicoAlbuquerqueNew MexicoUSA
| | - Jessie Sanford
- Department of PsychiatryWashington University in St LouisSt. LouisMissouriUSA
- NeuroGenomics and Informatics CenterWashington University in St LouisMissouriUSA
| | - Chia‐Ling Phuah
- NeuroGenomics and Informatics CenterWashington University in St LouisMissouriUSA
- Department of NeurologyWashington University in St LouisSt. LouisMissouriUSA
| | - Jason Hassenstab
- Department of PsychologyWashington University in St LouisSt. LouisMissouriUSA
| | - Carlos Cruchaga
- Department of PsychiatryWashington University in St LouisSt. LouisMissouriUSA
- NeuroGenomics and Informatics CenterWashington University in St LouisMissouriUSA
| |
Collapse
|
50
|
Dincer A, Chen CD, McKay NS, Koenig LN, McCullough A, Flores S, Keefe SJ, Schultz SA, Feldman RL, Joseph-Mathurin N, Hornbeck RC, Cruchaga C, Schindler SE, Holtzman DM, Morris JC, Fagan AM, Benzinger TLS, Gordon BA. APOE ε4 genotype, amyloid-β, and sex interact to predict tau in regions of high APOE mRNA expression. Sci Transl Med 2022; 14:eabl7646. [PMID: 36383681 PMCID: PMC9912474 DOI: 10.1126/scitranslmed.abl7646] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The apolipoprotein E (APOE) ε4 allele is strongly linked with cerebral β-amyloidosis, but its relationship with tauopathy is less established. We investigated the relationship between APOE ε4 carrier status, regional amyloid-β (Aβ), magnetic resonance imaging (MRI) volumetrics, tau positron emission tomography (PET), APOE messenger RNA (mRNA) expression maps, and cerebrospinal fluid phosphorylated tau (CSF ptau181). Three hundred fifty participants underwent imaging, and 270 had ptau181. We used computational models to evaluate the main effect of APOE ε4 carrier status on regional neuroimaging values and then the interaction of ε4 status and global Aβ on regional tau PET and brain volumes as well as CSF ptau181. Separately, we also examined the additional interactive influence of sex. We found that, for the same degree of Aβ burden, APOE ε4 carriers showed greater tau PET signal relative to noncarriers in temporal regions, but no interaction was present for MRI volumes or CSF ptau181. This potentiation of tau aggregation irrespective of sex occurred in brain regions with high APOE mRNA expression, suggesting local vulnerabilities to tauopathy. There were greater effects of APOE genotype in females, although the interactive sex effects did not strongly mirror mRNA expression. Pathology is not homogeneously expressed throughout the brain but mirrors underlying biological patterns such as gene expression.
Collapse
Affiliation(s)
- Aylin Dincer
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Charles D Chen
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Nicole S McKay
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Lauren N Koenig
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Austin McCullough
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Shaney Flores
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Sarah J Keefe
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Stephanie A Schultz
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Rebecca L Feldman
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Nelly Joseph-Mathurin
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Russ C Hornbeck
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Carlos Cruchaga
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Suzanne E Schindler
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - David M Holtzman
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, Saint Louis, MO, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Anne M Fagan
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Tammie LS Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Brian A Gordon
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Psychological & Brain Sciences, Washington University, Saint Louis, MO, USA
| |
Collapse
|