1
|
Beckett LA, Saito N, Donohue MC, Harvey DJ. Contributions of the ADNI Biostatistics Core. Alzheimers Dement 2024; 20:7331-7339. [PMID: 39140601 PMCID: PMC11485306 DOI: 10.1002/alz.14159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 08/15/2024]
Abstract
The goal of the Biostatistics Core of the Alzheimer's Disease Neuroimaging Initiative (ADNI) has been to ensure that sound study designs and statistical methods are used to meet the overall goals of ADNI. We have supported the creation of a well-validated and well-curated longitudinal database of clinical and biomarker information on ADNI participants and helped to make this accessible and usable for researchers. We have developed a statistical methodology for characterizing the trajectories of clinical and biomarker change for ADNI participants across the spectrum from cognitively normal to dementia, including multivariate patterns and evidence for heterogeneity in cognitive aging. We have applied these methods and adapted them to improve clinical trial design. ADNI-4 will offer us a chance to help extend these efforts to a more diverse cohort with an even richer panel of biomarker data to support better knowledge of and treatment for Alzheimer's disease and related dementias. HIGHLIGHTS: The Alzheimer's Disease Neuroimaging Initiative (ADNI) Biostatistics Core provides study design and analytic support to ADNI investigators. Core members develop and apply novel statistical methodology to work with ADNI data and support clinical trial design. The Core contributes to the standardization, validation, and harmonization of biomarker data. The Core serves as a resource to the wider research community to address questions related to the data and study as a whole.
Collapse
Affiliation(s)
- Laurel A. Beckett
- Department of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | - Naomi Saito
- Department of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | - Michael C. Donohue
- Department of NeurologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Danielle J. Harvey
- Department of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | | |
Collapse
|
2
|
Guo Y, Chen SD, You J, Huang SY, Chen YL, Zhang Y, Wang LB, He XY, Deng YT, Zhang YR, Huang YY, Dong Q, Feng JF, Cheng W, Yu JT. Multiplex cerebrospinal fluid proteomics identifies biomarkers for diagnosis and prediction of Alzheimer's disease. Nat Hum Behav 2024; 8:2047-2066. [PMID: 38987357 DOI: 10.1038/s41562-024-01924-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 06/10/2024] [Indexed: 07/12/2024]
Abstract
Recent expansion of proteomic coverage opens unparalleled avenues to unveil new biomarkers of Alzheimer's disease (AD). Among 6,361 cerebrospinal fluid (CSF) proteins analysed from the ADNI database, YWHAG performed best in diagnosing both biologically (AUC = 0.969) and clinically (AUC = 0.857) defined AD. Four- (YWHAG, SMOC1, PIGR and TMOD2) and five- (ACHE, YWHAG, PCSK1, MMP10 and IRF1) protein panels greatly improved the accuracy to 0.987 and 0.975, respectively. Their superior performance was validated in an independent external cohort and in discriminating autopsy-confirmed AD versus non-AD, rivalling even canonical CSF ATN biomarkers. Moreover, they effectively predicted the clinical progression to AD dementia and were strongly associated with AD core biomarkers and cognitive decline. Synaptic, neurogenic and infectious pathways were enriched in distinct AD stages. Mendelian randomization did not support the significant genetic link between CSF proteins and AD. Our findings revealed promising high-performance biomarkers for AD diagnosis and prediction, with implications for clinical trials targeting different pathomechanisms.
Collapse
Affiliation(s)
- Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia You
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Shu-Yi Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Lin Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lin-Bo Wang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Xiao-Yu He
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yue-Ting Deng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Yuan Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Carmona-Iragui M, O'Connor A, Llibre-Guerra J, Lao P, Ashton NJ, Fortea J, Sánchez-Valle R. Clinical and research application of fluid biomarkers in autosomal dominant Alzheimer's disease and Down syndrome. EBioMedicine 2024; 108:105327. [PMID: 39366843 DOI: 10.1016/j.ebiom.2024.105327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/30/2024] [Accepted: 08/27/2024] [Indexed: 10/06/2024] Open
Abstract
Autosomal dominant Alzheimer's disease (ADAD) and Down syndrome (DS) constitute genetic forms of Alzheimer's disease (AD). The study of these forms has been crucial in understanding the biomarker changes and disease progression, notably in advancing our knowledge of the natural history of AD. However, some specific characteristics of biomarkers in genetically determined forms and, most importantly, the near full penetrance and predictability of disease onset lead to a very different context of use for biomarkers in these populations. This article delves into the similarities and differences in biomarker profiles between genetically determined AD and sporadic cases, discussing the implications for research and clinical practice. It also emphasizes the need to account for factors that may affect biomarker reliability differently in genetically determined AD. Enhancing our understanding of the disease will pave the way for more personalized therapeutic approaches for affected individuals.
Collapse
Affiliation(s)
- María Carmona-Iragui
- Sant Pau Memory Unit, Neurology Department. Hospital de la Santa Creu i Sant Pau- Biomedical Research Institute Sant Pau- Universitat Autònoma de Barcelona, Spain; Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Spain; Center of Biomedical Investigation Network for Neurodegenerative Diseases, CIBERNED, Spain.
| | - Antoinette O'Connor
- Department of Neurology, Tallaght University Hospital, Tallaght, Dublin 24, Ireland; Institute of Memory and Cognition, Tallaght University Hospital, Tallaght, Dublin 24, Ireland. antoinette.o'
| | - Jorge Llibre-Guerra
- Dominantly Inherited Alzheimer's Network Trials Unit, Department of Neurology, Washington University School of Medicine in St.Louis, USA.
| | - Patrick Lao
- G.H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10019, USA.
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK; NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK; Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway.
| | - Juan Fortea
- Sant Pau Memory Unit, Neurology Department. Hospital de la Santa Creu i Sant Pau- Biomedical Research Institute Sant Pau- Universitat Autònoma de Barcelona, Spain; Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Spain; Center of Biomedical Investigation Network for Neurodegenerative Diseases, CIBERNED, Spain.
| | - Raquel Sánchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Hospital Clínic de Barcelona, Fundació de Recerca Clínic Barcelona-IDIBAPS, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
4
|
Sewell KR, Doecke JD, Xiong C, Benzinger T, Masters CL, Laske C, Jucker M, Lopera F, Gordon BA, Llibre-Guerra J, Levin J, Huey ED, Hassenstab J, Schofield PR, Day GS, Fox NC, Chhatwal J, Ibanez L, Roh JH, Perrin R, Lee JH, Allegri RF, Supnet-Bell C, Berman SB, Daniels A, Noble J, Martins RN, Rainey-Smith S, Peiffer J, Gardener SL, Bateman RJ, Morris JC, McDade E, Erickson KI, Sohrabi HR, Brown BM. Longitudinal associations between exercise and biomarkers in autosomal dominant Alzheimer's disease. Alzheimers Dement 2024. [PMID: 39324510 DOI: 10.1002/alz.14270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/08/2024] [Accepted: 08/27/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION We investigated longitudinal associations between self-reported exercise and Alzheimer's disease (AD)-related biomarkers in individuals with autosomal dominant AD (ADAD) mutations. METHODS Participants were 308 ADAD mutation carriers aged 39.7 ± 10.8 years from the Dominantly Inherited Alzheimer's Network. Weekly exercise volume was measured via questionnaire and associations with brain volume (magnetic resonance imaging), cerebrospinal fluid biomarkers, and brain amyloid beta (Aβ) measured by positron emission tomography were investigated. RESULTS Greater volume of weekly exercise at baseline was associated with slower accumulation of brain Aβ at preclinical disease stages β = -0.16 [-0.23 to -0.08], and a slower decline in multiple brain regions including hippocampal volume β = 0.06 [0.03 to 0.08]. DISCUSSION Exercise is associated with more favorable profiles of AD-related biomarkers in individuals with ADAD mutations. Exercise may have therapeutic potential for delaying the onset of AD; however, randomized controlled trials are vital to determine a causal relationship before a clinical recommendation of exercise is implemented. HIGHLIGHTS Greater self-reported weekly exercise predicts slower declines in brain volume in autosomal dominant Alzheimer's disease (ADAD). Greater self-reported weekly exercise predicts slower accumulation of brain amyloid beta in ADAD. Associations varied depending on closeness to estimated symptom onset.
Collapse
Affiliation(s)
- Kelsey R Sewell
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia
| | - James D Doecke
- Australian E-Health Research Centre, CSIRO, Herston, Queensland, Australia
| | - Chengjie Xiong
- Washington University in St. Louis, St. Louis, Missouri, USA
| | | | - Colin L Masters
- The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Christoph Laske
- German Center for Neurodegenerative Diseases, Tubingen, Germany
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases, Tubingen, Germany
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Brian A Gordon
- Washington University in St. Louis, St. Louis, Missouri, USA
| | | | - Johannes Levin
- Department of Neurology, LMU University Hospital, LMU, Munich, Germany
- German Center for Neurodegenerative Diseases, Site Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Edward D Huey
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Jason Hassenstab
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Neurology, Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - 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
| | - Gregory S Day
- Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Jasmeer Chhatwal
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Laura Ibanez
- Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jee Hoon Roh
- Department of Neurology, Korea University Anam Hospital, Seoul, South Korea
- Department of Physiology, Korea University College of Medicine, Seoul, South Korea
| | - Richard Perrin
- Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jae-Hong Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ricardo F Allegri
- Cognitive Neurology Service of the FLENI Foundation, Foundation for Childhood Neurological Disorders, Cognitive Neurology, Neuropsychology and Neuropsychiatry Section (CONICET-FLENI), Buenos Aires, Argentina
| | | | - Sarah B Berman
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alisha Daniels
- Department of Neurology, Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - James Noble
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Ralph N Martins
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Alzheimer's Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
- Department of Biomedical Sciences, Macquarie University, North Ryde, New South Wales, Australia
| | - Stephanie Rainey-Smith
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Alzheimer's Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
| | - Jeremiah Peiffer
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia
| | - Samantha L Gardener
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Alzheimer's Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
| | | | - John C Morris
- Washington University in St. Louis, St. Louis, Missouri, USA
| | - Eric McDade
- Washington University in St. Louis, St. Louis, Missouri, USA
| | - Kirk I Erickson
- AdventHealth Research Institute, Neuroscience, Orlando, Florida, USA
| | - Hamid R Sohrabi
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Alzheimer's Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
- Department of Biomedical Sciences, Macquarie University, North Ryde, New South Wales, Australia
| | - Belinda M Brown
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Alzheimer's Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
| |
Collapse
|
5
|
Arafi P, Devkota S, Maesako M, Wolfe MS. Alzheimer-mutant γ-secretase complexes stall amyloid β-peptide production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.30.610520. [PMID: 39257787 PMCID: PMC11383658 DOI: 10.1101/2024.08.30.610520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Missense mutations in the amyloid precursor protein (APP) and presenilin-1 (PSEN1) cause early-onset familial Alzheimer's disease (FAD) and alter proteolytic production of secreted 38-to-43-residue amyloid β-peptides (Aβ) by the PSEN1-containing γ-secretase complex, ostensibly supporting the amyloid hypothesis of pathogenesis. However, proteolysis of APP substrate by γ-secretase is processive, involving initial endoproteolysis to produce long Aβ peptides of 48 or 49 residues followed by carboxypeptidase trimming in mostly tripeptide increments. We recently reported evidence that FAD mutations in APP and PSEN1 cause deficiencies in early steps in processive proteolysis of APP substrate C99 and that this results from stalled γ-secretase enzyme-substrate and/or enzyme-intermediate complexes. These stalled complexes triggered synaptic degeneration in a C. elegans model of FAD independently of Aβ production. Here we conducted full quantitative analysis of all proteolytic events on APP substrate by γ-secretase with six additional PSEN1 FAD mutations and found that all six are deficient in multiple processing steps. However, only one of these (F386S) was deficient in certain trimming steps but not in endoproteolysis. Fluorescence lifetime imaging microscopy in intact cells revealed that all six PSEN1 FAD mutations lead to stalled γ-secretase enzyme-substrate/intermediate complexes. The F386S mutation, however, does so only in Aβ-rich regions of the cells, not in C99-rich regions, consistent with the deficiencies of this mutant enzyme only in trimming of Aβ intermediates. These findings provide further evidence that FAD mutations lead stalled and stabilized γ-secretase enzyme-substrate and/or enzyme-intermediate complexes and are consistent with the stalled process rather than the products of γ-secretase proteolysis as the pathogenic trigger.
Collapse
Affiliation(s)
- Parnian Arafi
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, USA
| | - Sujan Devkota
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, USA
| | - Masato Maesako
- Alzheimer Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael S Wolfe
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
6
|
Zhang J, Zhang Y, Wang J, Xia Y, Zhang J, Chen L. Recent advances in Alzheimer's disease: Mechanisms, clinical trials and new drug development strategies. Signal Transduct Target Ther 2024; 9:211. [PMID: 39174535 PMCID: PMC11344989 DOI: 10.1038/s41392-024-01911-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/18/2024] [Accepted: 07/02/2024] [Indexed: 08/24/2024] Open
Abstract
Alzheimer's disease (AD) stands as the predominant form of dementia, presenting significant and escalating global challenges. Its etiology is intricate and diverse, stemming from a combination of factors such as aging, genetics, and environment. Our current understanding of AD pathologies involves various hypotheses, such as the cholinergic, amyloid, tau protein, inflammatory, oxidative stress, metal ion, glutamate excitotoxicity, microbiota-gut-brain axis, and abnormal autophagy. Nonetheless, unraveling the interplay among these pathological aspects and pinpointing the primary initiators of AD require further elucidation and validation. In the past decades, most clinical drugs have been discontinued due to limited effectiveness or adverse effects. Presently, available drugs primarily offer symptomatic relief and often accompanied by undesirable side effects. However, recent approvals of aducanumab (1) and lecanemab (2) by the Food and Drug Administration (FDA) present the potential in disrease-modifying effects. Nevertheless, the long-term efficacy and safety of these drugs need further validation. Consequently, the quest for safer and more effective AD drugs persists as a formidable and pressing task. This review discusses the current understanding of AD pathogenesis, advances in diagnostic biomarkers, the latest updates of clinical trials, and emerging technologies for AD drug development. We highlight recent progress in the discovery of selective inhibitors, dual-target inhibitors, allosteric modulators, covalent inhibitors, proteolysis-targeting chimeras (PROTACs), and protein-protein interaction (PPI) modulators. Our goal is to provide insights into the prospective development and clinical application of novel AD drugs.
Collapse
Affiliation(s)
- Jifa Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yinglu Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, TN, USA
| | - Yilin Xia
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxian Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lei Chen
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
7
|
Morris JC, Buckles VD. The role of the Alzheimer's Disease Neuroimaging Initiative in establishing the Dominantly Inherited Alzheimer Network. Alzheimers Dement 2024; 20:5789-5791. [PMID: 38958563 PMCID: PMC11350018 DOI: 10.1002/alz.14106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/06/2024] [Indexed: 07/04/2024]
Abstract
The Dominantly Inherited Alzheimer Network (DIAN) initially was funded by the National Institute on Aging (NIA) in 2008 and thus was able to adopt and incorporate the protocols developed by the Alzheimer's Disease Neuroimaging Initiative (ADNI) that had been established by the NIA in 2004. The use of ADNI protocols for DIAN neuroimaging studies and assays of biological fluids for Alzheimer disease (AD) biomarkers permitted examination of the hypothesis that autosomal dominant AD (ADAD), studied by DIAN, and "sporadic" late-onset AD (LOAD), studied by ADNI, shared the same pathobiological construct. In a collaborative effort, the longitudinal DIAN and ADNI databases were compared and the findings supported the conclusion that ADAD and LOAD share a similar pathophysiology. The importance of the DIAN study thus is amplified by its relevance to LOAD, as characterized by the "parent" ADNI program.
Collapse
Affiliation(s)
- John C. Morris
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSaint LouisMissouriUSA
| | - Virginia D. Buckles
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSaint LouisMissouriUSA
| |
Collapse
|
8
|
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
|
9
|
Pettersson M, Johnson DS, Humphrey JM, Am Ende CW, Butler TW, Dorff PH, Efremov IV, Evrard E, Green ME, Helal CJ, Kauffman GW, Mullins PB, Navaratnam T, O'Donnell CJ, O'Sullivan TJ, Patel NC, Stepan AF, Stiff CM, Subramanyam C, Trapa P, Tran TP, Vetelino BC, Yang E, Xie L, Pustilnik LR, Steyn SJ, Wood KM, Bales KR, Hajos-Korcsok E, Verhoest PR. Discovery of Clinical Candidate PF-06648671: A Potent γ-Secretase Modulator for the Treatment of Alzheimer's Disease. J Med Chem 2024; 67:10248-10262. [PMID: 38848667 DOI: 10.1021/acs.jmedchem.4c00580] [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: 06/09/2024]
Abstract
Herein, we describe the design and synthesis of γ-secretase modulator (GSM) clinical candidate PF-06648671 (22) for the treatment of Alzheimer's disease. A key component of the design involved a 2,5-cis-tetrahydrofuran (THF) linker to impart conformational rigidity and lock the compound into a putative bioactive conformation. This effort was guided using a pharmacophore model since crystallographic information was not available for the membrane-bound γ-secretase protein complex at the time of this work. PF-06648671 achieved excellent alignment of whole cell in vitro potency (Aβ42 IC50 = 9.8 nM) and absorption, distribution, metabolism, and excretion (ADME) parameters. This resulted in favorable in vivo pharmacokinetic (PK) profile in preclinical species, and PF-06648671 achieved a human PK profile suitable for once-a-day dosing. Furthermore, PF-06648671 was found to have favorable brain availability in rodent, which translated into excellent central exposure in human and robust reduction of amyloid β (Aβ) 42 in cerebrospinal fluid (CSF).
Collapse
Affiliation(s)
- Martin Pettersson
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, United States
| | - Douglas S Johnson
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, United States
| | - John M Humphrey
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | | | - Todd W Butler
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Peter H Dorff
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Ivan V Efremov
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Edelweiss Evrard
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, United States
| | - Michael E Green
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, United States
| | - Christopher J Helal
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Gregory W Kauffman
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, United States
| | - Patrick B Mullins
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Thayalan Navaratnam
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | | | - Theresa J O'Sullivan
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Nandini C Patel
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, United States
| | - Antonia F Stepan
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, United States
| | - Cory M Stiff
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | | | - Patrick Trapa
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, United States
| | - Tuan P Tran
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Beth Cooper Vetelino
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Eddie Yang
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Longfei Xie
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Leslie R Pustilnik
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Stefanus J Steyn
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, United States
| | - Kathleen M Wood
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Kelly R Bales
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, United States
| | - Eva Hajos-Korcsok
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, United States
| | - Patrick R Verhoest
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
10
|
Levin J, Baiardi S, Quadalti C, Rossi M, Mammana A, Vöglein J, Bernhardt A, Perrin RJ, Jucker M, Preische O, Hofmann A, Höglinger GU, Cairns NJ, Franklin EE, Chrem P, Cruchaga C, Berman SB, Chhatwal JP, Daniels A, Day GS, Ryan NS, Goate AM, Gordon BA, Huey ED, Ibanez L, Karch CM, Lee J, Llibre‐Guerra J, Lopera F, Masters CL, Morris JC, Noble JM, Renton AE, Roh JH, Frosch MP, Keene CD, McLean C, Sanchez‐Valle R, Schofield PR, Supnet‐Bell C, Xiong C, Giese A, Hansson O, Bateman RJ, McDade E, Parchi P. α-Synuclein seed amplification assay detects Lewy body co-pathology in autosomal dominant Alzheimer's disease late in the disease course and dependent on Lewy pathology burden. Alzheimers Dement 2024; 20:4351-4365. [PMID: 38666355 PMCID: PMC11180868 DOI: 10.1002/alz.13818] [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: 11/30/2023] [Revised: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 06/01/2024]
Abstract
INTRODUCTION Amyloid beta and tau pathology are the hallmarks of sporadic Alzheimer's disease (AD) and autosomal dominant AD (ADAD). However, Lewy body pathology (LBP) is found in ≈ 50% of AD and ADAD brains. METHODS Using an α-synuclein seed amplification assay (SAA) in cerebrospinal fluid (CSF) from asymptomatic (n = 26) and symptomatic (n = 27) ADAD mutation carriers, including 12 with known neuropathology, we investigated the timing of occurrence and prevalence of SAA positive reactivity in ADAD in vivo. RESULTS No asymptomatic participant and only 11% (3/27) of the symptomatic patients tested SAA positive. Neuropathology revealed LBP in 10/12 cases, primarily affecting the amygdala or the olfactory areas. In the latter group, only the individual with diffuse LBP reaching the neocortex showed α-synuclein seeding activity in CSF in vivo. DISCUSSION Results suggest that in ADAD LBP occurs later than AD pathology and often as amygdala- or olfactory-predominant LBP, for which CSF α-synuclein SAA has low sensitivity. HIGHLIGHTS Cerebrospinal fluid (CSF) real-time quaking-induced conversion (RT-QuIC) detects misfolded α-synuclein in ≈ 10% of symptomatic autosomal dominant Alzheimer's disease (ADAD) patients. CSF RT-QuIC does not detect α-synuclein seeding activity in asymptomatic mutation carriers. Lewy body pathology (LBP) in ADAD mainly occurs as olfactory only or amygdala-predominant variants. LBP develops late in the disease course in ADAD. CSF α-synuclein RT-QuIC has low sensitivity for focal, low-burden LBP.
Collapse
Affiliation(s)
- Johannes Levin
- Department of NeurologyLMU University Hospital, LMU MunichMunichGermany
- German Center for Neurodegenerative DiseasesMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Simone Baiardi
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
| | - Corinne Quadalti
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
| | - Marcello Rossi
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
| | - Angela Mammana
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
| | - Jonathan Vöglein
- Department of NeurologyLMU University Hospital, LMU MunichMunichGermany
- German Center for Neurodegenerative DiseasesMunichGermany
| | - Alexander Bernhardt
- Department of NeurologyLMU University Hospital, LMU MunichMunichGermany
- German Center for Neurodegenerative DiseasesMunichGermany
| | - Richard J. Perrin
- Department of Pathology and ImmunologyWashington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
- Hertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Oliver Preische
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
- Hertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Anna Hofmann
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
- Hertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Günter U. Höglinger
- Department of NeurologyLMU University Hospital, LMU MunichMunichGermany
- German Center for Neurodegenerative DiseasesMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Nigel J. Cairns
- Living Systems InstituteFaculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Erin E. Franklin
- Department of Pathology and ImmunologyWashington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | | | - Carlos Cruchaga
- Department of PsychiatryWashington University School of MedicineSaint LouisMissouriUSA
| | | | - Jasmeer P. Chhatwal
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Alisha Daniels
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Gregory S. Day
- Department of NeurologyMayo Clinic in FloridaJacksonvilleFloridaUSA
| | - Natalie S. Ryan
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - Alison M. Goate
- Department of Genetics & Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Brian A. Gordon
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Edward D. Huey
- Butler HospitalBrown Center for Alzheimer's Disease ResearchAlpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Laura Ibanez
- Department of PsychiatryWashington University School of MedicineSaint LouisMissouriUSA
| | - Celeste M. Karch
- Department of PsychiatryWashington University School of MedicineSaint LouisMissouriUSA
| | - Jae‐Hong Lee
- Department of NeurologyAsan Medical CenterSeoulSouth Korea
| | - Jorge Llibre‐Guerra
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Francisco Lopera
- Grupo de Neurosciencias de Antioquia, Sede de Investigación Universitaria SIUMedellínColombia
| | - Colin L. Masters
- Florey Institute and The University of MelbourneMelbourneVictoriaAustralia
| | - John C. Morris
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | - James M. Noble
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and the Aging Brain, and GH Sergievsky Center, Columbia UniversityNew YorkNew YorkUSA
| | - Alan E. Renton
- Department of Genetics and Genomic Sciences and Nash Family Dept of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Jee Hoon Roh
- Departments of Neurology and PhysiologyKorea University College of MedicineSeoulSouth Korea
| | - Matthew P. Frosch
- MassGeneral Institute for Neurodegenerative Diseases, Neuropathology Service, Massachusetts General HospitalBostonMassachusettsUSA
| | - C. Dirk Keene
- Department of Laboratory Medicine and PathologyUniversity of WashingtonSeattleWashingtonUSA
| | - Catriona McLean
- Department of Anatomical PathologyAlfredHealthMelbourneVictoriaAustralia
| | - Raquel Sanchez‐Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Service of Neurology, Hospital Clinic de Barcelona, FRCB‐IDIBAPSBarcelonaSpain
| | - Peter R. Schofield
- Neuroscience Research AustraliaSydneyNew South WalesAustralia
- School of Medical SciencesUniversity of New South WalesSydneyNew South WalesAustralia
| | - Charlene Supnet‐Bell
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Chengjie Xiong
- Division of BiostatisticsWashington University School of MedicineSaint LouisMissouriUSA
| | | | - Oskar Hansson
- Clinical Memory Research UnitDepartment of Clinical Sciences MalmöFaculty of Medicine, Lund UniversityLundSweden
- Memory ClinicSkåne University HospitalLundSweden
| | - Randall J. Bateman
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Eric McDade
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | | | - Piero Parchi
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
| |
Collapse
|
11
|
Teipel S, Grazia A, Dyrba M, Grothe MJ, Pomara N. Basal forebrain volume and metabolism in carriers of the Colombian mutation for autosomal dominant Alzheimer's disease. Sci Rep 2024; 14:11268. [PMID: 38760448 PMCID: PMC11101449 DOI: 10.1038/s41598-024-60799-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/26/2024] [Indexed: 05/19/2024] Open
Abstract
We aimed to study atrophy and glucose metabolism of the cholinergic basal forebrain in non-demented mutation carriers for autosomal dominant Alzheimer's disease (ADAD). We determined the level of evidence for or against atrophy and impaired metabolism of the basal forebrain in 167 non-demented carriers of the Colombian PSEN1 E280A mutation and 75 age- and sex-matched non-mutation carriers of the same kindred using a Bayesian analysis framework. We analyzed baseline MRI, amyloid PET, and FDG-PET scans of the Alzheimer's Prevention Initiative ADAD Colombia Trial. We found moderate evidence against an association of carrier status with basal forebrain volume (Bayes factor (BF10) = 0.182). We found moderate evidence against a difference of basal forebrain metabolism (BF10 = 0.167). There was only inconclusive evidence for an association between basal forebrain volume and delayed memory and attention (BF10 = 0.884 and 0.184, respectively), and between basal forebrain volume and global amyloid load (BF10 = 2.1). Our results distinguish PSEN1 E280A mutation carriers from sporadic AD cases in which cholinergic involvement of the basal forebrain is already detectable in the preclinical and prodromal stages. This indicates an important difference between ADAD and sporadic AD in terms of pathogenesis and potential treatment targets.
Collapse
Affiliation(s)
- Stefan Teipel
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Gehlsheimer Str. 20, 18147, Rostock, Germany.
- Department of Psychosomatic Medicine, University Medicine Rostock, Gehlsheimer Str. 20, 18147, Rostock, Germany.
| | - Alice Grazia
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Gehlsheimer Str. 20, 18147, Rostock, Germany
| | - Martin Dyrba
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Gehlsheimer Str. 20, 18147, Rostock, Germany
| | - Michel J Grothe
- CIEN Foundation/Queen Sofia Foundation Alzheimer Center, Madrid, Spain
| | - Nunzio Pomara
- Geriatric Psychiatry Division, Nathan Kline Institute/Department of Psychiatry and Pathology, NYU Grossman School of Medicine, Orangeburg, NY, USA
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Korczyn AD, Grinberg LT. Is Alzheimer disease a disease? Nat Rev Neurol 2024; 20:245-251. [PMID: 38424454 DOI: 10.1038/s41582-024-00940-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2024] [Indexed: 03/02/2024]
Abstract
Dementia, a prevalent condition among older individuals, has profound societal implications. Extensive research has resulted in no cure for what is perceived as the most common dementing illness: Alzheimer disease (AD). AD is defined by specific brain abnormalities - amyloid-β plaques and tau protein neurofibrillary tangles - that are proposed to actively influence the neurodegenerative process. However, conclusive evidence of amyloid-β toxicity is lacking, the mechanisms leading to the accumulation of plaques and tangles are unknown, and removing amyloid-β has not halted neurodegeneration. So, the question remains, are we making progress towards a solution? The complexity of AD is underscored by numerous genetic and environmental risk factors, and diverse clinical presentations, suggesting that AD is more akin to a syndrome than to a traditional disease, with its pathological manifestation representing a convergence of pathogenic pathways. Therefore, a solution requires a multifaceted approach over a single 'silver bullet'. Improved recognition and classification of conditions that converge in plaques and tangle accumulation and their treatment requires the use of multiple strategies simultaneously.
Collapse
Affiliation(s)
- Amos D Korczyn
- Departments of Neurology, Physiology and Pharmacology, Tel Aviv University, Tel Aviv, Israel.
| | - Lea T Grinberg
- Departments of Neurology and Pathology, UCSF, San Francisco, CA, USA
- Global Brain Health Institute, UCSF, San Francisco, CA, USA
- Department of Pathology, University of Sao Paulo Medical School, Sao Paulo, Brazil
| |
Collapse
|
15
|
Therriault J, Schindler SE, Salvadó G, Pascoal TA, Benedet AL, Ashton NJ, Karikari TK, Apostolova L, Murray ME, Verberk I, Vogel JW, La Joie R, Gauthier S, Teunissen C, Rabinovici GD, Zetterberg H, Bateman RJ, Scheltens P, Blennow K, Sperling R, Hansson O, Jack CR, Rosa-Neto P. Biomarker-based staging of Alzheimer disease: rationale and clinical applications. Nat Rev Neurol 2024; 20:232-244. [PMID: 38429551 DOI: 10.1038/s41582-024-00942-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2024] [Indexed: 03/03/2024]
Abstract
Disease staging, whereby the spatial extent and load of brain pathology are used to estimate the severity of Alzheimer disease (AD), is pivotal to the gold-standard neuropathological diagnosis of AD. Current in vivo diagnostic frameworks for AD are based on abnormal concentrations of amyloid-β and tau in the cerebrospinal fluid or on PET scans, and breakthroughs in molecular imaging have opened up the possibility of in vivo staging of AD. Focusing on the key principles of disease staging shared across several areas of medicine, this Review highlights the potential for in vivo staging of AD to transform our understanding of preclinical AD, refine enrolment criteria for trials of disease-modifying therapies and aid clinical decision-making in the era of anti-amyloid therapeutics. We provide a state-of-the-art review of recent biomarker-based AD staging systems and highlight their contributions to the understanding of the natural history of AD. Furthermore, we outline hypothetical frameworks to stage AD severity using more accessible fluid biomarkers. In addition, by applying amyloid PET-based staging to recently published anti-amyloid therapeutic trials, we highlight how biomarker-based disease staging frameworks could illustrate the numerous pathological changes that have already taken place in individuals with mildly symptomatic AD. Finally, we discuss challenges related to the validation and standardization of disease staging and provide a forward-looking perspective on potential clinical applications.
Collapse
Affiliation(s)
- Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, Quebec, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada.
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andréa Lessa Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- NIHR Biomedical Research Centre, South London and Maudsley NHS Foundation, London, UK
| | - Thomas K Karikari
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Liana Apostolova
- Department of Neurology, University of Indiana School of Medicine, Indianapolis, IN, USA
| | | | - Inge Verberk
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Jacob W Vogel
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Clinical Sciences, Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Charlotte Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Philip Scheltens
- Alzheimer Centre Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Reisa Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | | | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
16
|
Devkota S, Zhou R, Nagarajan V, Maesako M, Do H, Noorani A, Overmeyer C, Bhattarai S, Douglas JT, Saraf A, Miao Y, Ackley BD, Shi Y, Wolfe MS. Familial Alzheimer mutations stabilize synaptotoxic γ-secretase-substrate complexes. Cell Rep 2024; 43:113761. [PMID: 38349793 PMCID: PMC10941010 DOI: 10.1016/j.celrep.2024.113761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/19/2023] [Accepted: 01/24/2024] [Indexed: 02/15/2024] Open
Abstract
Mutations that cause familial Alzheimer's disease (FAD) are found in amyloid precursor protein (APP) and presenilin, the catalytic component of γ-secretase, that together produce amyloid β-peptide (Aβ). Nevertheless, whether Aβ is the primary disease driver remains controversial. We report here that FAD mutations disrupt initial proteolytic events in the multistep processing of APP substrate C99 by γ-secretase. Cryoelectron microscopy reveals that a substrate mimetic traps γ-secretase during the transition state, and this structure aligns with activated enzyme-substrate complex captured by molecular dynamics simulations. In silico simulations and in cellulo fluorescence microscopy support stabilization of enzyme-substrate complexes by FAD mutations. Neuronal expression of C99 and/or presenilin-1 in Caenorhabditis elegans leads to synaptic loss only with FAD-mutant transgenes. Designed mutations that stabilize the enzyme-substrate complex and block Aβ production likewise led to synaptic loss. Collectively, these findings implicate the stalled process-not the products-of γ-secretase cleavage of substrates in FAD pathogenesis.
Collapse
Affiliation(s)
- Sujan Devkota
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, USA
| | - Rui Zhou
- Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | | | - Masato Maesako
- Alzheimer Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hung Do
- Center for Computational Biology, University of Kansas, Lawrence, KS, USA
| | - Arshad Noorani
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, USA
| | - Caitlin Overmeyer
- Graduate Program in Neurosciences, University of Kansas, Lawrence, KS, USA
| | - Sanjay Bhattarai
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, USA
| | - Justin T Douglas
- Nuclear Magnetic Resonance Core Lab, University of Kansas, Lawrence, KS, USA
| | - Anita Saraf
- Mass Spectrometry and Analytical Proteomic Laboratory, University of Kansas, Lawrence, KS, USA
| | - Yinglong Miao
- Center for Computational Biology, University of Kansas, Lawrence, KS, USA; Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Brian D Ackley
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Yigong Shi
- Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China; Westlake Laboratory of Life Science and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, and Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China
| | - Michael S Wolfe
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, USA; Graduate Program in Neurosciences, University of Kansas, Lawrence, KS, USA.
| |
Collapse
|
17
|
De Bastiani MA, Bellaver B, Carello-Collar G, Zimmermann M, Kunach P, Lima-Filho RA, Forner S, Martini AC, Pascoal TA, Lourenco MV, Rosa-Neto P, Zimmer ER. Cross-species comparative hippocampal transcriptomics in Alzheimer's disease. iScience 2024; 27:108671. [PMID: 38292167 PMCID: PMC10824791 DOI: 10.1016/j.isci.2023.108671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 07/11/2023] [Accepted: 12/05/2023] [Indexed: 02/01/2024] Open
Abstract
Alzheimer's disease (AD) is a multifactorial pathology, with most cases having a sporadic origin. Recently, knock-in (KI) mouse models, such as the novel humanized amyloid-β (hAβ)-KI, have been developed to better resemble sporadic human AD. METHODS Here, we compared hippocampal publicly available transcriptomic profiles of transgenic (5xFAD and APP/PS1) and KI (hAβ-KI) mouse models with early- (EOAD) and late- (LOAD) onset AD patients. RESULTS The three mouse models presented more Gene Ontology biological processes terms and enriched signaling pathways in common with LOAD than with EOAD individuals. Experimental validation of consistently dysregulated genes revealed five altered in mice (SLC11A1, S100A6, CD14, CD33, and C1QB) and three in humans (S100A6, SLC11A1, and KCNK). Finally, we identified 17 transcription factors potentially acting as master regulators of AD. CONCLUSION Our cross-species analyses revealed that the three mouse models presented a remarkable similarity to LOAD, with the hAβ-KI being the more specific one.
Collapse
Affiliation(s)
- Marco Antônio De Bastiani
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Health Basic Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, State of Rio Grande do Sul 90035-003, Brazil
| | - Bruna Bellaver
- Department of Psychiatry, School of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Giovanna Carello-Collar
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Health Basic Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, State of Rio Grande do Sul 90035-003, Brazil
| | - Maria Zimmermann
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Québec H3A 1A1, Canada
- Translational Neuroimaging Laboratory, McGill University, Montréal, Québec H4H 1R3, Canada
| | - Peter Kunach
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Québec H3A 1A1, Canada
- Translational Neuroimaging Laboratory, McGill University, Montréal, Québec H4H 1R3, Canada
- Douglas Hospital Research Centre, Montreal, Québec H4H 1R3, Canada
| | - Ricardo A.S. Lima-Filho
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, State of Rio de Janeiro 21941-902, Brazil
| | - Stefania Forner
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA 92697, USA
| | - Alessandra Cadete Martini
- Department of Pathology & Laboratory Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Tharick A. Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Mychael V. Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, State of Rio de Janeiro 21941-902, Brazil
| | - Pedro Rosa-Neto
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Québec H3A 1A1, Canada
- Translational Neuroimaging Laboratory, McGill University, Montréal, Québec H4H 1R3, Canada
- Douglas Hospital Research Centre, Montreal, Québec H4H 1R3, Canada
| | - Eduardo R. Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Health Basic Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, State of Rio Grande do Sul 90035-003, Brazil
- Department of Pharmacology, ICBS, UFRGS, Porto Alegre, State of Rio Grande do Sul 90035-003, Brazil
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Department of Pharmacology, ICBS, UFRGS, Porto Alegre, State of Rio Grande do Sul 90035-003, Brazil
- Brain Institute of Rio Grande Do Sul, Pontifical Catholic University of Rio Grande Do Sul, Porto Alegre, State of Rio Grande do Sul 90610-000, Brazil
| |
Collapse
|
18
|
Wolfe MS. γ-Secretase: once and future drug target for Alzheimer's disease. Expert Opin Drug Discov 2024; 19:5-8. [PMID: 37915204 PMCID: PMC10872755 DOI: 10.1080/17460441.2023.2277350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023]
Affiliation(s)
- Michael S. Wolfe
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS 66049 USA
| |
Collapse
|
19
|
Veitch DP, Weiner MW, Miller M, Aisen PS, Ashford MA, Beckett LA, Green RC, Harvey D, Jack CR, Jagust W, Landau SM, Morris JC, Nho KT, Nosheny R, Okonkwo O, Perrin RJ, Petersen RC, Rivera Mindt M, Saykin A, Shaw LM, Toga AW, Tosun D. The Alzheimer's Disease Neuroimaging Initiative in the era of Alzheimer's disease treatment: A review of ADNI studies from 2021 to 2022. Alzheimers Dement 2024; 20:652-694. [PMID: 37698424 PMCID: PMC10841343 DOI: 10.1002/alz.13449] [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: 04/24/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 09/13/2023]
Abstract
The Alzheimer's Disease Neuroimaging Initiative (ADNI) aims to improve Alzheimer's disease (AD) clinical trials. Since 2006, ADNI has shared clinical, neuroimaging, and cognitive data, and biofluid samples. We used conventional search methods to identify 1459 publications from 2021 to 2022 using ADNI data/samples and reviewed 291 impactful studies. This review details how ADNI studies improved disease progression understanding and clinical trial efficiency. Advances in subject selection, detection of treatment effects, harmonization, and modeling improved clinical trials and plasma biomarkers like phosphorylated tau showed promise for clinical use. Biomarkers of amyloid beta, tau, neurodegeneration, inflammation, and others were prognostic with individualized prediction algorithms available online. Studies supported the amyloid cascade, emphasized the importance of neuroinflammation, and detailed widespread heterogeneity in disease, linked to genetic and vascular risk, co-pathologies, sex, and resilience. Biological subtypes were consistently observed. Generalizability of ADNI results is limited by lack of cohort diversity, an issue ADNI-4 aims to address by enrolling a diverse cohort.
Collapse
Affiliation(s)
- Dallas P. Veitch
- Department of Veterans Affairs Medical CenterNorthern California Institute for Research and Education (NCIRE)San FranciscoCaliforniaUSA
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
| | - Michael W. Weiner
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of MedicineUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of Psychiatry and Behavioral SciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Melanie Miller
- Department of Veterans Affairs Medical CenterNorthern California Institute for Research and Education (NCIRE)San FranciscoCaliforniaUSA
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
| | - Paul S. Aisen
- Alzheimer's Therapeutic Research InstituteUniversity of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Miriam A. Ashford
- Department of Veterans Affairs Medical CenterNorthern California Institute for Research and Education (NCIRE)San FranciscoCaliforniaUSA
| | - Laurel A. Beckett
- Division of BiostatisticsDepartment of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | - Robert C. Green
- Division of GeneticsDepartment of MedicineBrigham and Women's HospitalBroad Institute Ariadne Labs and Harvard Medical SchoolBostonMassachusettsUSA
| | - Danielle Harvey
- Division of BiostatisticsDepartment of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | | | - William Jagust
- Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Susan M. Landau
- Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - John C. Morris
- Knight Alzheimer's Disease Research CenterWashington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
- Department of Pathology and ImmunologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Kwangsik T. Nho
- Department of Radiology and Imaging Sciences and the Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for Computational Biology and BioinformaticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Rachel Nosheny
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Psychiatry and Behavioral SciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Ozioma Okonkwo
- Wisconsin Alzheimer's Disease Research Center and Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Richard J. Perrin
- Knight Alzheimer's Disease Research CenterWashington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
- Department of Pathology and ImmunologyWashington University School of MedicineSaint LouisMissouriUSA
| | | | - Monica Rivera Mindt
- Department of PsychologyLatin American and Latino Studies InstituteAfrican and African American StudiesFordham UniversityNew YorkNew YorkUSA
- Department of NeurologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Andrew Saykin
- Department of Radiology and Imaging Sciences and the Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine and the PENN Alzheimer's Disease Research CenterCenter for Neurodegenerative ResearchPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Arthur W. Toga
- Laboratory of Neuro ImagingInstitute of Neuroimaging and InformaticsKeck School of Medicine of University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Duygu Tosun
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | | |
Collapse
|
20
|
Millar PR, Gordon BA, Wisch JK, Schultz SA, Benzinger TL, Cruchaga C, Hassenstab JJ, Ibanez L, Karch C, Llibre-Guerra JJ, Morris JC, Perrin RJ, Supnet-Bell C, Xiong C, Allegri RF, Berman SB, Chhatwal JP, Chrem Mendez PA, Day GS, Hofmann A, Ikeuchi T, Jucker M, Lee JH, Levin J, Lopera F, Niimi Y, Sánchez-González VJ, Schofield PR, Sosa-Ortiz AL, Vöglein J, Bateman RJ, Ances BM, McDade EM. Advanced structural brain aging in preclinical autosomal dominant Alzheimer disease. Mol Neurodegener 2023; 18:98. [PMID: 38111006 PMCID: PMC10729487 DOI: 10.1186/s13024-023-00688-3] [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: 07/03/2023] [Accepted: 11/28/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND "Brain-predicted age" estimates biological age from complex, nonlinear features in neuroimaging scans. The brain age gap (BAG) between predicted and chronological age is elevated in sporadic Alzheimer disease (AD), but is underexplored in autosomal dominant AD (ADAD), in which AD progression is highly predictable with minimal confounding age-related co-pathology. METHODS We modeled BAG in 257 deeply-phenotyped ADAD mutation-carriers and 179 non-carriers from the Dominantly Inherited Alzheimer Network using minimally-processed structural MRI scans. We then tested whether BAG differed as a function of mutation and cognitive status, or estimated years until symptom onset, and whether it was associated with established markers of amyloid (PiB PET, CSF amyloid-β-42/40), phosphorylated tau (CSF and plasma pTau-181), neurodegeneration (CSF and plasma neurofilament-light-chain [NfL]), and cognition (global neuropsychological composite and CDR-sum of boxes). We compared BAG to other MRI measures, and examined heterogeneity in BAG as a function of ADAD mutation variants, APOE ε4 carrier status, sex, and education. RESULTS Advanced brain aging was observed in mutation-carriers approximately 7 years before expected symptom onset, in line with other established structural indicators of atrophy. BAG was moderately associated with amyloid PET and strongly associated with pTau-181, NfL, and cognition in mutation-carriers. Mutation variants, sex, and years of education contributed to variability in BAG. CONCLUSIONS We extend prior work using BAG from sporadic AD to ADAD, noting consistent results. BAG associates well with markers of pTau, neurodegeneration, and cognition, but to a lesser extent, amyloid, in ADAD. BAG may capture similar signal to established MRI measures. However, BAG offers unique benefits in simplicity of data processing and interpretation. Thus, results in this unique ADAD cohort with few age-related confounds suggest that brain aging attributable to AD neuropathology can be accurately quantified from minimally-processed MRI.
Collapse
Affiliation(s)
- Peter R Millar
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA.
| | - Brian A Gordon
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Julie K Wisch
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Stephanie A Schultz
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Tammie Ls Benzinger
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Jason J Hassenstab
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Laura Ibanez
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
- NeuroGenomics & Informatics Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Celeste Karch
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | | | - John C Morris
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Richard J Perrin
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Chengjie Xiong
- Department of Biostatistics, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Sarah B Berman
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Gregory S Day
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | - Anna Hofmann
- German Center for Neurodegenerative Diseases (DZNE), 72076, Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076, Tübingen, Germany
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases (DZNE), 72076, Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076, Tübingen, Germany
| | - Jae-Hong Lee
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - 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
| | | | - Yoshiki Niimi
- Unit for Early and Exploratory Clinical Development, The University of Tokyo Hospital, Bunkyo-Ku, Tokyo, Japan
| | - Victor J Sánchez-González
- Departamento de Clínicas, CUALTOS, Universidad de Guadalajara, Tepatitlán de Morelos, Jalisco, México
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, NSW, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Ana Luisa Sosa-Ortiz
- Instituto Nacional de Neurologia y Neurocirugía MVS, CDMX, Ciudad de México, Mexico
| | - Jonathan Vöglein
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Randall J Bateman
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Beau M Ances
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Eric M McDade
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
21
|
van der Ende EL, In ‘t Veld SGJG, Hanskamp I, van der Lee S, Dijkstra JIR, Hok-A-Hin YS, Blujdea ER, van Swieten JC, Irwin DJ, Chen-Plotkin A, Hu WT, Lemstra AW, Pijnenburg YAL, van der Flier WM, del Campo M, Teunissen CE, Vermunt L. CSF proteomics in autosomal dominant Alzheimer's disease highlights parallels with sporadic disease. Brain 2023; 146:4495-4507. [PMID: 37348871 PMCID: PMC10629764 DOI: 10.1093/brain/awad213] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 06/24/2023] Open
Abstract
Autosomal dominant Alzheimer's disease (ADAD) offers a unique opportunity to study pathophysiological changes in a relatively young population with few comorbidities. A comprehensive investigation of proteome changes occurring in ADAD could provide valuable insights into AD-related biological mechanisms and uncover novel biomarkers and therapeutic targets. Furthermore, ADAD might serve as a model for sporadic AD, but in-depth proteome comparisons are lacking. We aimed to identify dysregulated CSF proteins in ADAD and determine the degree of overlap with sporadic AD. We measured 1472 proteins in CSF of PSEN1 or APP mutation carriers (n = 22) and age- and sex-matched controls (n = 20) from the Amsterdam Dementia Cohort using proximity extension-based immunoassays (PEA). We compared protein abundance between groups with two-sided t-tests and identified enriched biological pathways. Using the same protein panels in paired plasma samples, we investigated correlations between CSF proteins and their plasma counterparts. Finally, we compared our results with recently published PEA data from an international cohort of sporadic AD (n = 230) and non-AD dementias (n = 301). All statistical analyses were false discovery rate-corrected. We detected 66 differentially abundant CSF proteins (65 increased, 1 decreased) in ADAD compared to controls (q < 0.05). The most strongly upregulated proteins (fold change >1.8) were related to immunity (CHIT1, ITGB2, SMOC2), cytoskeletal structure (MAPT, NEFL) and tissue remodelling (TMSB10, MMP-10). Significant CSF-plasma correlations were found for the upregulated proteins SMOC2 and LILR1B. Of the 66 differentially expressed proteins, 36 had been measured previously in the sporadic dementias cohort, 34 of which (94%) were also significantly upregulated in sporadic AD, with a strong correlation between the fold changes of these proteins in both cohorts (rs = 0.730, P < 0.001). Twenty-nine of the 36 proteins (81%) were also upregulated among non-AD patients with suspected AD co-pathology. This CSF proteomics study demonstrates substantial biochemical similarities between ADAD and sporadic AD, suggesting involvement of the same biological processes. Besides known AD-related proteins, we identified several relatively novel proteins, such as TMSB10, MMP-10 and SMOC2, which have potential as novel biomarkers. With shared pathophysiological CSF changes, ADAD study findings might be translatable to sporadic AD, which could greatly expedite therapy development.
Collapse
Affiliation(s)
- Emma L van der Ende
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Sjors G J G In ‘t Veld
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Iris Hanskamp
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Sven van der Lee
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Janna I R Dijkstra
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yanaika S Hok-A-Hin
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Elena R Blujdea
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - John C van Swieten
- Alzheimer Center and Department of Neurology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William T Hu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Afina W Lemstra
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Epidemiology and Data Science, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Marta del Campo
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28003 Madrid, Spain
- Barcelonabeta Brain Research Center (BBRC), Pasqual Maragall Foundation, 08005 Barcelona, Spain
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Lisa Vermunt
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
22
|
Adhikari S, Qiao Y, Singer M, Sagare A, Jiang X, Shi Y, Ringman JM, Kashani AH. Retinotopic degeneration of the retina and optic tracts in autosomal dominant Alzheimer's disease. Alzheimers Dement 2023; 19:5103-5113. [PMID: 37102308 PMCID: PMC10603214 DOI: 10.1002/alz.13100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/28/2023]
Abstract
INTRODUCTION We investigated the correlation between retinal thickness and optic tract integrity in subjects with autosomal dominant Alzheimer's disease (ADAD) causing mutations. METHODS Retinal thicknesses and diffusion tensor images (DTI) were obtained using optical coherence tomography and magnetic resonance imaging, respectively. The association between retinal thickness and DTI measures was adjusted for age, sex, retinotopy, and correlation between eyes. RESULTS Optic tract mean diffusivity and axial diffusivity were negatively correlated with retinotopically defined ganglion cell inner plexiform thickness (GCIPL). Fractional anisotropy was negatively correlated with retinotopically defined retinal nerve fiber layer thickness. There was no correlation between outer nuclear layer (ONL) thickness and any DTI measure. DISCUSSION In ADAD, GCIPL thickness is significantly associated with retinotopic optic tract DTI measures even in minimally symptomatic subjects. Similar associations were not present with ONL thickness or when ignoring retinotopy. We provide in vivo evidence for optic tract changes resulting from ganglion cell pathology in ADAD.
Collapse
Affiliation(s)
- Suman Adhikari
- Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yuchuan Qiao
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Maxwell Singer
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Abhay Sagare
- Zilkha Neurogenetics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Department of Neurology, Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Xuejuan Jiang
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Yonggang Shi
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - John M Ringman
- Department of Neurology, Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Amir H Kashani
- Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
23
|
Jagust WJ, Teunissen CE, DeCarli C. The complex pathway between amyloid β and cognition: implications for therapy. Lancet Neurol 2023; 22:847-857. [PMID: 37454670 DOI: 10.1016/s1474-4422(23)00128-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/11/2023] [Accepted: 03/27/2023] [Indexed: 07/18/2023]
Abstract
For decades, the hypothesis that brain deposition of the amyloid β protein initiates Alzheimer's disease has dominated research and clinical trials. Targeting amyloid β is starting to produce therapeutic benefit, although whether amyloid-lowering drugs will be widely and meaningfully effective is still unclear. Despite extensive in-vivo biomarker evidence in humans showing the importance of an amyloid cascade that drives cognitive decline, the amyloid hypothesis does not fully account for the complexity of late-life cognitive impairment. Multiple brain pathological changes, inflammation, and host factors of resilience might also be involved in contributing to the development of dementia. This variability suggests that the benefits of lowering amyloid β might depend on how strongly an amyloid pathway is manifest in an individual in relation to other coexisting pathophysiological processes. A new approach to research and treatment, which fully considers the multiple factors that drive cognitive decline, is necessary.
Collapse
Affiliation(s)
- William J Jagust
- School of Public Health, and Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA.
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Charles DeCarli
- Department of Neurology, University of California, Davis, CA, USA
| |
Collapse
|
24
|
Events in the brain during the evolution of Alzheimer's disease. Nat Med 2023; 29:1916-1917. [PMID: 37550418 DOI: 10.1038/s41591-023-02477-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
|
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
|
Johnson ECB, Bian S, Haque RU, Carter EK, Watson CM, Gordon BA, Ping L, Duong DM, Epstein MP, McDade E, Barthélemy NR, Karch CM, Xiong C, Cruchaga C, Perrin RJ, Wingo AP, Wingo TS, Chhatwal JP, Day GS, Noble JM, Berman SB, Martins R, Graff-Radford NR, Schofield PR, Ikeuchi T, Mori H, Levin J, Farlow M, Lah JJ, Haass C, Jucker M, Morris JC, Benzinger TLS, Roberts BR, Bateman RJ, Fagan AM, Seyfried NT, Levey AI. Cerebrospinal fluid proteomics define the natural history of autosomal dominant Alzheimer's disease. Nat Med 2023; 29:1979-1988. [PMID: 37550416 PMCID: PMC10427428 DOI: 10.1038/s41591-023-02476-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/27/2023] [Indexed: 08/09/2023]
Abstract
Alzheimer's disease (AD) pathology develops many years before the onset of cognitive symptoms. Two pathological processes-aggregation of the amyloid-β (Aβ) peptide into plaques and the microtubule protein tau into neurofibrillary tangles (NFTs)-are hallmarks of the disease. However, other pathological brain processes are thought to be key disease mediators of Aβ plaque and NFT pathology. How these additional pathologies evolve over the course of the disease is currently unknown. Here we show that proteomic measurements in autosomal dominant AD cerebrospinal fluid (CSF) linked to brain protein coexpression can be used to characterize the evolution of AD pathology over a timescale spanning six decades. SMOC1 and SPON1 proteins associated with Aβ plaques were elevated in AD CSF nearly 30 years before the onset of symptoms, followed by changes in synaptic proteins, metabolic proteins, axonal proteins, inflammatory proteins and finally decreases in neurosecretory proteins. The proteome discriminated mutation carriers from noncarriers before symptom onset as well or better than Aβ and tau measures. Our results highlight the multifaceted landscape of AD pathophysiology and its temporal evolution. Such knowledge will be critical for developing precision therapeutic interventions and biomarkers for AD beyond those associated with Aβ and tau.
Collapse
Affiliation(s)
- Erik C B Johnson
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Shijia Bian
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Rafi U Haque
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | - E Kathleen Carter
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Caroline M Watson
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Brian A Gordon
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Lingyan Ping
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Duc M Duong
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael P Epstein
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric McDade
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | | | - Celeste M Karch
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Chengjie Xiong
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
- Division of Biostatistics, Washington University in St Louis, St Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, 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
| | - Aliza P Wingo
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, USA
- Division of Mental Health, Atlanta VA Medical Center, Atlanta, GA, USA
| | - Thomas S Wingo
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Jasmeer P Chhatwal
- Massachusetts General and Brigham & Women's Hospitals, Harvard Medical School, Boston, MA, USA
| | - Gregory S Day
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | - James M Noble
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, and GH Sergievsky Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Sarah B Berman
- Departments of Neurology and Clinical and Translational Science, Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ralph Martins
- Edith Cowan University, Perth, Western Australia, Australia
| | | | - 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
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hiroshi Mori
- Osaka Metropolitan University Medical School, Nagaoka Sutoku University, Nagaoka, Japan
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - James J Lah
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Metabolic Biochemistry, Biomedical Center (BMC), Ludwig-Maximilians University, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - 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
| | - Blaine R Roberts
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Randall J Bateman
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Anne M Fagan
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Nicholas T Seyfried
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Allan I Levey
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
27
|
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
|
28
|
Aguillon D, Langella S, Chen Y, Sanchez J, Su Y, Vila-Castelar C, Vasquez D, Zetterberg H, Hansson O, Dage JL, Janelidze S, Chen K, Fox-Fuller JT, Aduen P, Martinez JE, Garcia G, Baena A, Guzman C, Johnson K, Sperling RA, Blennow K, Reiman EM, Lopera F, Quiroz YT. Plasma p-tau217 predicts in vivo brain pathology and cognition in autosomal dominant Alzheimer's disease. Alzheimers Dement 2023; 19:2585-2594. [PMID: 36571821 PMCID: PMC10271963 DOI: 10.1002/alz.12906] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Plasma-measured tau phosphorylated at threonine 217 (p-tau217) is a potential non-invasive biomarker of Alzheimer's disease (AD). We investigated whether plasma p-tau217 predicts subsequent cognition and positron emission tomography (PET) markers of pathology in autosomal dominant AD. METHODS We analyzed baseline levels of plasma p-tau217 and its associations with amyloid PET, tau PET, and word list delayed recall measured 7.61 years later in non-demented age- and education-matched presenilin-1 E280A carriers (n = 24) and non-carrier (n = 20) family members. RESULTS Carriers had higher plasma p-tau217 levels than non-carriers. Baseline plasma p-tau217 was associated with subsequent amyloid and tau PET pathology levels and cognitive function. DISCUSSION Our findings suggest that plasma p-tau217 predicts subsequent brain pathological burden and memory performance in presenilin-1 E280A carriers. These results provide support for plasma p-tau217 as a minimally invasive diagnostic and prognostic biomarker for AD, with potential utility in clinical practice and trials. HIGHLIGHTS Non-demented presenilin-1 E280A carriers have higher plasma tau phosphorylated at threonine 217 (p-tau217) than do age-matched non-carriers. Higher baseline p-tau217 is associated with greater future amyloid positron emission tomography (PET) pathology burden. Higher baseline p-tau217 is associated with greater future tau PET pathology burden. Higher baseline p-tau217 is associated with worse future memory performance.
Collapse
Affiliation(s)
- David Aguillon
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | | | | | - Justin Sanchez
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yi Su
- Banner Alzheimer’s Institute, Phoenix, AZ, USA
| | | | - Daniel Vasquez
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Oskar Hansson
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
| | - Jeffrey L. Dage
- Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Kewei Chen
- Banner Alzheimer’s Institute, Phoenix, AZ, USA
| | - Joshua T. Fox-Fuller
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychological and Brain Sciences, Boston University, Boston, USA
| | - Paula Aduen
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jairo E. Martinez
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychological and Brain Sciences, Boston University, Boston, USA
| | - Gloria Garcia
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Ana Baena
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Claudia Guzman
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Keith Johnson
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Reisa A. Sperling
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | | | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Yakeel T. Quiroz
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|