1
|
Gillis G, Bhalerao G, Blane J, Mitchell R, Pretorius PM, McCracken C, Nichols TE, Smith SM, Miller KL, Alfaro‐Almagro F, Raymont V, Martos L, Mackay CE, Griffanti L. From Big Data to the Clinic: Methodological and Statistical Enhancements to Implement the UK Biobank Imaging Framework in a Memory Clinic. Hum Brain Mapp 2025; 46:e70151. [PMID: 39969115 PMCID: PMC11837031 DOI: 10.1002/hbm.70151] [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: 08/14/2024] [Revised: 11/27/2024] [Accepted: 01/18/2025] [Indexed: 02/20/2025] Open
Abstract
The analysis tools and statistical methods used in large neuroimaging research studies differ from those applied in clinical contexts, making it unclear whether these techniques can be translated to a memory clinic setting. The Oxford Brain Health Clinic (OBHC) was established in 2020 to bridge this gap between research studies and memory clinics. We optimised the UK Biobank imaging framework for the memory clinic setting by integrating enhanced quality control (QC) processes (MRIQC, QUAD, and DSE decomposition) and supplementary dementia-informed analyses (lobar volumes, NBM volumes, WMH classification, PSMD, cortical diffusion MRI metrics, and tract volumes) into the analysis pipeline. We explored associations between resultant imaging-derived phenotypes (IDPs) and clinical phenotypes in the OBHC patient population (N = 213), applying hierarchical FDR correction to account for multiple testing. 14%-24% of scans were flagged by automated QC tools, but upon visual inspection, only 0%-2.4% of outputs were excluded. The pipeline successfully generated 5683 IDPs aligned with UK Biobank and 110 IDPs targeted towards dementia-related changes. We replicated established associations and found novel associations between brain metrics and age, cognition, and dementia-related diagnoses. The imaging protocol is feasible, acceptable, and yields high-quality data that is usable for both clinical and research purposes. We validated the use of this methodology in a real-world memory clinic population, which demonstrates the potential of this enhanced pipeline to bridge the gap between big data studies and clinical settings.
Collapse
Affiliation(s)
- Grace Gillis
- Department of Psychiatry, Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative NeuroimagingUniversity of OxfordOxfordUK
- Oxford Health NHS Foundation TrustOxfordUK
| | - Gaurav Bhalerao
- Department of Psychiatry, Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative NeuroimagingUniversity of OxfordOxfordUK
- Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging, FMRIBUniversity of OxfordOxfordUK
| | - Jasmine Blane
- Department of Psychiatry, Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative NeuroimagingUniversity of OxfordOxfordUK
- Oxford Health NHS Foundation TrustOxfordUK
| | | | - Pieter M. Pretorius
- Department of NeuroradiologyOxford University Hospitals NHS Foundation TrustOxfordUK
| | - Celeste McCracken
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Thomas E. Nichols
- Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging, FMRIBUniversity of OxfordOxfordUK
| | - Stephen M. Smith
- Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging, FMRIBUniversity of OxfordOxfordUK
| | - Karla L. Miller
- Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging, FMRIBUniversity of OxfordOxfordUK
| | - Fidel Alfaro‐Almagro
- Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging, FMRIBUniversity of OxfordOxfordUK
| | - Vanessa Raymont
- Department of Psychiatry, Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative NeuroimagingUniversity of OxfordOxfordUK
- Oxford Health NHS Foundation TrustOxfordUK
| | - Lola Martos
- Department of Psychiatry, Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative NeuroimagingUniversity of OxfordOxfordUK
- Oxford Health NHS Foundation TrustOxfordUK
| | - Clare E. Mackay
- Department of Psychiatry, Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative NeuroimagingUniversity of OxfordOxfordUK
- Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging, FMRIBUniversity of OxfordOxfordUK
| | - Ludovica Griffanti
- Department of Psychiatry, Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative NeuroimagingUniversity of OxfordOxfordUK
- Oxford Health NHS Foundation TrustOxfordUK
- Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging, FMRIBUniversity of OxfordOxfordUK
| |
Collapse
|
2
|
Appleton J, Finn Q, Zanotti-Fregonara P, Yu M, Faridar A, Nakawah MO, Zarate C, Carrillo MC, Dickerson BC, Rabinovici GD, Apostolova LG, Masdeu JC, Pascual B. Brain inflammation co-localizes highly with tau in mild cognitive impairment due to early-onset Alzheimer's disease. Brain 2025; 148:119-132. [PMID: 39013020 PMCID: PMC11706285 DOI: 10.1093/brain/awae234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/27/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024] Open
Abstract
Brain inflammation, with an increased density of microglia and macrophages, is an important component of Alzheimer's disease and a potential therapeutic target. However, it is incompletely characterized, particularly in patients whose disease begins before the age of 65 years and, thus, have few co-pathologies. Inflammation has been usefully imaged with translocator protein (TSPO) PET, but most inflammation PET tracers cannot image subjects with a low-binder TSPO rs6971 genotype. In an important development, participants with any TSPO genotype can be imaged with a novel tracer, 11C-ER176, that has a high binding potential and a more favourable metabolite profile than other TSPO tracers currently available. We applied 11C-ER176 to detect brain inflammation in mild cognitive impairment (MCI) caused by early-onset Alzheimer's disease. Furthermore, we sought to correlate the brain localization of inflammation, volume loss, elevated amyloid-β (Aβ)and tau. We studied brain inflammation in 25 patients with early-onset amnestic MCI (average age 59 ± 4.5 years, 10 female) and 23 healthy controls (average age 65 ± 6.0 years, 12 female), both groups with a similar proportion of all three TSPO-binding affinities. 11C-ER176 total distribution volume (VT), obtained with an arterial input function, was compared across patients and controls using voxel-wise and region-wise analyses. In addition to inflammation PET, most MCI patients had Aβ (n = 23) and tau PET (n = 21). For Aβ and tau tracers, standard uptake value ratios were calculated using cerebellar grey matter as region of reference. Regional correlations among the three tracers were determined. Data were corrected for partial volume effect. Cognitive performance was studied with standard neuropsychological tools. In MCI caused by early-onset Alzheimer's disease, there was inflammation in the default network, reaching statistical significance in precuneus and lateral temporal and parietal association cortex bilaterally, and in the right amygdala. Topographically, inflammation co-localized most strongly with tau (r = 0.63 ± 0.24). This correlation was higher than the co-localization of Aβ with tau (r = 0.55 ± 0.25) and of inflammation with Aβ (0.43 ± 0.22). Inflammation co-localized least with atrophy (-0.29 ± 0.26). These regional correlations could be detected in participants with any of the three rs6971 TSPO polymorphisms. Inflammation in Alzheimer's disease-related regions correlated with impaired cognitive scores. Our data highlight the importance of inflammation, a potential therapeutic target, in the Alzheimer's disease process. Furthermore, they support the notion that, as shown in experimental tissue and animal models, the propagation of tau in humans is associated with brain inflammation.
Collapse
Affiliation(s)
- Johanna Appleton
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Quentin Finn
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | | | - Meixiang Yu
- Cyclotron and Radiopharmaceutical Core, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Alireza Faridar
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Mohammad O Nakawah
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Carlos Zarate
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Maria C Carrillo
- Medical & Scientific Relations Division, Alzheimer's Association, Chicago, IL 60603, USA
| | | | - Gil D Rabinovici
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Liana G Apostolova
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Joseph C Masdeu
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Belen Pascual
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| |
Collapse
|
3
|
Maure-Blesa L, Rodríguez-Baz I, Carmona-Iragui M, Fortea J. What Can We Learn About Alzheimer's Disease from People with Down Syndrome? Curr Top Behav Neurosci 2025; 69:197-226. [PMID: 39509049 DOI: 10.1007/7854_2024_546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Down syndrome (DS) is the most frequent cause of intellectual disability of genetic origin, estimated to affect about 1 in 700 babies born worldwide (CDC 2023). In Europe and the United States, current estimates indicate a population prevalence of 5.6 and 6.7 per 10,000 individuals, respectively, which translates to more than 200,000 people in the United States, more than 400,000 people in Europe, and approximately six million worldwide. Advances in healthcare and the treatment of accompanying conditions have significantly prolonged the lifespan of those with DS over the past 50 years. Consequently, there is a pressing need to address the challenges associated with ageing among this population, with Alzheimer's disease (AD) being the primary concern. In this chapter, we will review the significance of studying this population to understand AD biology, the insights gained on AD in DS (DSAD), and how this knowledge can help us understand the AD not only in DS but also in the general population. We will conclude by exploring the objectives that remain to be accomplished.
Collapse
Affiliation(s)
- Lucia Maure-Blesa
- Sant Pau Memory Unit, Department of Neurology, Facultad de Medicina, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Iñigo Rodríguez-Baz
- Sant Pau Memory Unit, Department of Neurology, Facultad de Medicina, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Maria Carmona-Iragui
- Sant Pau Memory Unit, Department of Neurology, Facultad de Medicina, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Facultad de Medicina, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain.
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain.
| |
Collapse
|
4
|
Belder CRS, Boche D, Nicoll JAR, Jaunmuktane Z, Zetterberg H, Schott JM, Barkhof F, Fox NC. Brain volume change following anti-amyloid β immunotherapy for Alzheimer's disease: amyloid-removal-related pseudo-atrophy. Lancet Neurol 2024; 23:1025-1034. [PMID: 39304242 DOI: 10.1016/s1474-4422(24)00335-1] [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: 05/13/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 09/22/2024]
Abstract
Progressive cerebral volume loss on MRI is a hallmark of Alzheimer's disease and has been widely used as an outcome measure in clinical trials, with the prediction that disease-modifying treatments would slow loss. However, in trials of anti-amyloid immunotherapy, the participants who received treatment had excess volume loss. Explanations for this observation range from reduction of amyloid β plaque burden and related inflammatory changes through to treatment-induced toxicity. The excess volume changes are characteristic of only those immunotherapies that achieve amyloid β lowering; are compatible with plaque removal; and evidence to date does not suggest an association with harmful effects. Based on the current evidence, we suggest that these changes can be described as amyloid-removal-related pseudo-atrophy. Better understanding of the causes and consequences of these changes is important to enable informed decisions about treatments. Patient-level analyses of data from the trials are urgently needed, along with longitudinal follow-up and neuroimaging data, to determine the long-term trajectory of these volume changes and their clinical correlates. Post-mortem examination of cerebral tissue from treated patients and evaluation of potential correlation with antemortem neuroimaging findings are key priorities.
Collapse
Affiliation(s)
- Christopher R S Belder
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK; UK Dementia Research Institute at UCL, University College London, London, UK; Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - James A R Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Zane Jaunmuktane
- Department of Clinical and Movement Neurosciences, Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Henrik Zetterberg
- UK Dementia Research Institute at UCL, University College London, London, UK; 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 Queen Square Institute of Neurology, University College London, London, UK; Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong Special Administrative Region, China; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Jonathan M Schott
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, London, UK
| | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK; UK Dementia Research Institute at UCL, University College London, London, UK.
| |
Collapse
|
5
|
Sun P, He Z, Li A, Yang J, Zhu Y, Cai Y, Ma T, Ma S, Guo T. Spatial and temporal patterns of cortical mean diffusivity in Alzheimer's disease and suspected non-Alzheimer's disease pathophysiology. Alzheimers Dement 2024; 20:7048-7061. [PMID: 39132849 PMCID: PMC11485315 DOI: 10.1002/alz.14176] [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: 04/12/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION The spatial and temporal patterns of cortical mean diffusivity (cMD), as well as its association with Alzheimer's disease (AD) and suspected non-Alzheimer's pathophysiology (SNAP), are not yet fully understood. METHODS We compared baseline (n = 617) and longitudinal changes (n = 421) of cMD, cortical thickness, and gray matter volume and their relations to vascular risk factors, amyloid beta (Aβ), and tau positron emission tomography (PET), and longitudinal cognitive decline in Aβ PET negative and positive older adults. RESULTS cMD increases were more sensitive to detecting brain structural alterations than cortical thinning and gray matter atrophy. Tau-related cMD increases partially mediated Aβ-related cognitive decline in AD, whereas vascular disease-related increased cMD levels substantially mediated age-related cognitive decline in SNAP. DISCUSSION These findings revealed the dynamic changes of microstructural and macrostructural indicators and their associations with AD and SNAP, providing novel insights into understanding upstream and downstream events of cMD in neurodegenerative disease. HIGHLIGHTS Cortical mean diffusivity (cMD) was more sensitive to detecting structural changes than macrostructural factors. Tau-related cMD increases partially mediated amyloid beta-related cognitive decline in Alzheimer's disease (AD). White matter hyperintensity-related higher cMD mainly explained the age-related cognitive decline in suspected non-Alzheimer's pathophysiology (SNAP). cMD may assist in tracking earlier neurodegenerative signs in AD and SNAP.
Collapse
Affiliation(s)
- Pan Sun
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
- Tsinghua Shenzhen International Graduate School (SIGS)Tsinghua UniversityShenzhenChina
| | - Zhengbo He
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
| | - Anqi Li
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
| | - Jie Yang
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
| | - Yalin Zhu
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
| | - Yue Cai
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
| | - Ting Ma
- School of Electronic and Information EngineeringHarbin Institute of Technology (Shenzhen)ShenzhenChina
| | - Shaohua Ma
- Tsinghua Shenzhen International Graduate School (SIGS)Tsinghua UniversityShenzhenChina
| | - Tengfei Guo
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
- Institute of Biomedical EngineeringPeking University Shenzhen Graduate SchoolShenzhenChina
| | | |
Collapse
|
6
|
Daniels AJ, McDade E, Llibre-Guerra JJ, Xiong C, Perrin RJ, Ibanez L, Supnet-Bell C, Cruchaga C, Goate A, Renton AE, Benzinger TL, Gordon BA, Hassenstab J, Karch C, Popp B, Levey A, Morris J, Buckles V, Allegri RF, Chrem P, Berman SB, Chhatwal JP, Farlow MR, Fox NC, Day GS, Ikeuchi T, Jucker M, Lee JH, Levin J, Lopera F, Takada L, Sosa AL, Martins R, Mori H, Noble JM, Salloway S, Huey E, Rosa-Neto P, Sánchez-Valle R, Schofield PR, Roh JH, Bateman RJ. 15 Years of Longitudinal Genetic, Clinical, Cognitive, Imaging, and Biochemical Measures in DIAN. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.08.24311689. [PMID: 39148846 PMCID: PMC11326320 DOI: 10.1101/2024.08.08.24311689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
This manuscript describes and summarizes the Dominantly Inherited Alzheimer Network Observational Study (DIAN Obs), highlighting the wealth of longitudinal data, samples, and results from this human cohort study of brain aging and a rare monogenic form of Alzheimer's disease (AD). DIAN Obs is an international collaborative longitudinal study initiated in 2008 with support from the National Institute on Aging (NIA), designed to obtain comprehensive and uniform data on brain biology and function in individuals at risk for autosomal dominant AD (ADAD). ADAD gene mutations in the amyloid protein precursor (APP), presenilin 1 (PSEN1), or presenilin 2 (PSEN2) genes are deterministic causes of ADAD, with virtually full penetrance, and a predictable age at symptomatic onset. Data and specimens collected are derived from full clinical assessments, including neurologic and physical examinations, extensive cognitive batteries, structural and functional neuro-imaging, amyloid and tau pathological measures using positron emission tomography (PET), flurordeoxyglucose (FDG) PET, cerebrospinal fluid and blood collection (plasma, serum, and whole blood), extensive genetic and multi-omic analyses, and brain donation upon death. This comprehensive evaluation of the human nervous system is performed longitudinally in both mutation carriers and family non-carriers, providing one of the deepest and broadest evaluations of the human brain across decades and through AD progression. These extensive data sets and samples are available for researchers to address scientific questions on the human brain, aging, and AD.
Collapse
Affiliation(s)
- Alisha J. Daniels
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Eric McDade
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | | | - Chengjie Xiong
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Richard J. Perrin
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Laura Ibanez
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | | | - Carlos Cruchaga
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Alison Goate
- Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Alan E. Renton
- Icahn School of Medicine at Mount Sinai, New York, NY USA
| | | | - Brian A. Gordon
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Jason Hassenstab
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Celeste Karch
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Brent Popp
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Allan Levey
- Goizueta Alzheimer’s Disease Research Center, Emory University, Atlanta, GA, USA
| | - John Morris
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Virginia Buckles
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | | | - Patricio Chrem
- Institute of Neurological Research FLENI, Buenos Aires, Argentina
| | | | - Jasmeer P. Chhatwal
- Massachusetts General and Brigham & Women’s Hospitals, Harvard Medical School, Boston MA, USA
| | | | - Nick C. Fox
- UK Dementia Research Institute at University College London, London, United Kingdom
- University College London, London, United Kingdom
| | | | - Takeshi Ikeuchi
- Brain Research Institute, Niigata University, Niigata, Japan
| | - Mathias Jucker
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- DZNE, German Center for Neurodegenerative Diseases, Tübingen, Germany
| | | | - Johannes Levin
- DZNE, German Center for Neurodegenerative Diseases, Munich, Germany
- Ludwig-Maximilians-Universität München, Munich, Germany
| | | | | | - Ana Luisa Sosa
- Instituto Nacional de Neurologia y Neurocirugla Innn, Mexico City, Mexico
| | - Ralph Martins
- Edith Cowan University, Western Australia, Australia
| | | | - James M. Noble
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Department of Neurology, and GH Sergievsky Center, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Edward Huey
- Brown University, Butler Hospital, Providence, RI, USA
| | - Pedro Rosa-Neto
- Centre de Recherche de L’hopital Douglas and McGill University, Montreal, Quebec
| | - Raquel Sánchez-Valle
- Hospital Clínic de Barcelona. IDIBAPS. University of Barcelona, Barcelona, Spain
| | - Peter R. Schofield
- Neuroscience Research Australia, Sydney, NSW, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Jee Hoon Roh
- Korea University, Korea University Anam Hospital, Seoul, South Korea
| | | | | |
Collapse
|
7
|
Chaudhary S, Roy A, Summers C, Ahles T, Li CSR, Chao HH. Androgen deprivation increases frontopolar cortical thickness in prostate cancer patients: an effect of early neurodegeneration? Am J Cancer Res 2024; 14:3652-3664. [PMID: 39113873 PMCID: PMC11301281 DOI: 10.62347/wola8904] [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: 04/03/2024] [Accepted: 07/15/2024] [Indexed: 08/10/2024] Open
Abstract
Androgen deprivation therapy (ADT) has been associated with adverse effects on the brain. ADT leads to altered testosterone levels that may affect brain morphology as well as cognition. Considering the reliability of cortical thickness (CT) as a marker of cognitive and brain changes, e.g., in Alzheimer's disease, we assessed the impacts of ADT on CT and working memory. Thirty men with non-metastatic prostate cancer receiving ADT and 32 patients not receiving ADT (controls or CON), matched in age and years of education, participated in N-back task and quality-of-life (QoL) assessments as well as brain imaging at baseline and prospectively at 6 months. Imaging data were processed with published routines to estimate CT and the results of a group by time flexible factorial analysis were evaluated at a corrected threshold. ADT and CON did not differ in N-back performance or QoL across time points. Relative to CON, patients receiving ADT showed significantly higher frontopolar cortex (FPC) CT at 6-month follow-up vs. baseline. Follow-up vs. baseline FPC CT change correlated negatively with changes in 2-back correct response rate and in testosterone levels across all participants. In mediation analysis, FPC CT change mediated the association between testosterone level change and 2-back accuracy rate change. Increases in FPC CT following 6 months of ADT may reflect early neurodegenerative changes in response to androgen deprivation. While no significant impact on working memory or QoL was observed over 6 months, further research of longer duration of treatment is warranted to unravel the full spectrum of cognitive and neural consequences of ADT in prostate cancer patients.
Collapse
Affiliation(s)
- Shefali Chaudhary
- Department of Psychiatry, Yale University School of MedicineNew Haven, CT, USA
| | - Alicia Roy
- VA Connecticut Healthcare SystemWest Haven, CT, USA
| | | | - Tim Ahles
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer CenterNew York, NY, USA
| | - Chiang-Shan R Li
- Department of Psychiatry, Yale University School of MedicineNew Haven, CT, USA
- Department of Neuroscience, Yale University School of MedicineNew Haven, CT, USA
- Interdepartmental Neuroscience Program, Yale University School of MedicineNew Haven, CT, USA
- Wu Tsai Institute, Yale UniversityNew Haven, CT, USA
| | - Herta H Chao
- VA Connecticut Healthcare SystemWest Haven, CT, USA
- Department of Medicine and Yale Comprehensive Cancer Center, Yale University School of MedicineNew Haven, CT, USA
| |
Collapse
|
8
|
Wearn A, Tremblay SA, Tardif CL, Leppert IR, Gauthier CJ, Baracchini G, Hughes C, Hewan P, Tremblay-Mercier J, Rosa-Neto P, Poirier J, Villeneuve S, Schmitz TW, Turner GR, Spreng RN. Neuromodulatory subcortical nucleus integrity is associated with white matter microstructure, tauopathy and APOE status. Nat Commun 2024; 15:4706. [PMID: 38830849 PMCID: PMC11148077 DOI: 10.1038/s41467-024-48490-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 05/01/2024] [Indexed: 06/05/2024] Open
Abstract
The neuromodulatory subcortical nuclei within the isodendritic core (IdC) are the earliest sites of tauopathy in Alzheimer's disease (AD). They project broadly throughout the brain's white matter. We investigated the relationship between IdC microstructure and whole-brain white matter microstructure to better understand early neuropathological changes in AD. Using multiparametric quantitative magnetic resonance imaging we observed two covariance patterns between IdC and white matter microstructure in 133 cognitively unimpaired older adults (age 67.9 ± 5.3 years) with familial risk for AD. IdC integrity related to 1) whole-brain neurite density, and 2) neurite orientation dispersion in white matter tracts known to be affected early in AD. Pattern 2 was associated with CSF concentration of phosphorylated-tau, indicating AD specificity. Apolipoprotein-E4 carriers expressed both patterns more strongly than non-carriers. IdC microstructure variation is reflected in white matter, particularly in AD-affected tracts, highlighting an early mechanism of pathological development.
Collapse
Affiliation(s)
- Alfie Wearn
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, H3A 2B4, QC, Canada.
| | - Stéfanie A Tremblay
- Department of Physics, Concordia University, Montreal, H4B 1R6, QC, Canada
- Montreal Heart Institute, Montreal, H1T 1C8, QC, Canada
- School of Health, Concordia University, Montreal, H4B 1R6, QC, Canada
| | - Christine L Tardif
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, H3A 2B4, QC, Canada
- McConnell Brain Imaging Centre, McGill University, Montreal, H3A 2B4, QC, Canada
- Department of Biomedical Engineering, McGill University, McGill, H3A 2B4, QC, Canada
| | - Ilana R Leppert
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, H3A 2B4, QC, Canada
- McConnell Brain Imaging Centre, McGill University, Montreal, H3A 2B4, QC, Canada
| | - Claudine J Gauthier
- Department of Physics, Concordia University, Montreal, H4B 1R6, QC, Canada
- Montreal Heart Institute, Montreal, H1T 1C8, QC, Canada
- School of Health, Concordia University, Montreal, H4B 1R6, QC, Canada
| | - Giulia Baracchini
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, H3A 2B4, QC, Canada
| | - Colleen Hughes
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, H3A 2B4, QC, Canada
| | - Patrick Hewan
- Department of Psychology, York University, Toronto, M3J 1P3, ON, Canada
| | | | - Pedro Rosa-Neto
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, H3A 2B4, QC, Canada
- McConnell Brain Imaging Centre, McGill University, Montreal, H3A 2B4, QC, Canada
- Douglas Mental Health University Institute-Research Center, Verdun, H4H 1R3, QC, Canada
| | - Judes Poirier
- Douglas Mental Health University Institute-Research Center, Verdun, H4H 1R3, QC, Canada
- Department of Psychiatry, McGill University, Montreal, H3A 1A1, QC, Canada
| | - Sylvia Villeneuve
- McConnell Brain Imaging Centre, McGill University, Montreal, H3A 2B4, QC, Canada
- Douglas Mental Health University Institute-Research Center, Verdun, H4H 1R3, QC, Canada
- Department of Psychiatry, McGill University, Montreal, H3A 1A1, QC, Canada
| | - Taylor W Schmitz
- Department of Physiology & Pharmacology, Western Institute for Neuroscience, Western University, London, N6A 5C1, ON, Canada
| | - Gary R Turner
- Department of Psychology, York University, Toronto, M3J 1P3, ON, Canada
| | - R Nathan Spreng
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, H3A 2B4, QC, Canada.
- McConnell Brain Imaging Centre, McGill University, Montreal, H3A 2B4, QC, Canada.
- Douglas Mental Health University Institute-Research Center, Verdun, H4H 1R3, QC, Canada.
- Department of Psychiatry, McGill University, Montreal, H3A 1A1, QC, Canada.
| |
Collapse
|
9
|
Pardo J, Montal V, Campabadal A, Oltra J, Uribe C, Roura I, Bargalló N, Martí MJ, Compta Y, Iranzo A, Fortea J, Junqué C, Segura B. Cortical Macro- and Microstructural Changes in Parkinson's Disease with Probable Rapid Eye Movement Sleep Behavior Disorder. Mov Disord 2024; 39:814-824. [PMID: 38456361 DOI: 10.1002/mds.29761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/17/2024] [Accepted: 02/16/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Evidence regarding cortical atrophy patterns in Parkinson's disease (PD) with probable rapid eye movement sleep behavior disorder (RBD) (PD-pRBD) remains scarce. Cortical mean diffusivity (cMD), as a novel imaging biomarker highly sensitive to detecting cortical microstructural changes in different neurodegenerative diseases, has not been investigated in PD-pRBD yet. OBJECTIVES The aim was to investigate cMD as a sensitive measure to identify subtle cortical microstructural changes in PD-pRBD and its relationship with cortical thickness (CTh). METHODS Twenty-two PD-pRBD, 31 PD without probable RBD (PD-nonpRBD), and 28 healthy controls (HC) were assessed using 3D T1-weighted and diffusion-weighted magnetic resonance imaging on a 3-T scanner and neuropsychological testing. Measures of cortical brain changes were obtained through cMD and CTh. Two-class group comparisons of a general linear model were performed (P < 0.05). Cohen's d effect size for both approaches was computed. RESULTS PD-pRBD patients showed higher cMD than PD-nonpRBD patients in the left superior temporal, superior frontal, and precentral gyri, precuneus cortex, as well as in the right middle frontal and postcentral gyri and paracentral lobule (d > 0.8), whereas CTh did not detect significant differences. PD-pRBD patients also showed increased bilateral posterior cMD in comparison with HCs (d > 0.8). These results partially overlapped with CTh results (0.5 < d < 0.8). PD-nonpRBD patients showed no differences in cMD when compared with HCs but showed cortical thinning in the left fusiform gyrus and lateral occipital cortex bilaterally (d > 0.5). CONCLUSIONS cMD may be more sensitive than CTh displaying significant cortico-structural differences between PD subgroups, indicating this imaging biomarker's utility in studying early cortical changes in PD. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Grants
- PID2020-114640GB-I00/AEI/10.13039/501100011033 Ministerio de Economía y Competitividad
- H2020-SC1-BHC-2018-2020/GA 965422 European Union's Horizon 2020, "MES-CoBraD"
- FI18/00275 Instituto de Salud Carlos III
- IIBSP-DOW-2020-151 Departament de Salut, Generalitat de Catalunya, Fundación Tatiana Pérez de Guzmán el Bueno
- PRE2018-086675 Ministerio de Ciencia, Innovación y Universidades
- PI20/01473 Fondo de Investigaciones Sanitario, Carlos III Health Institute
- SGR 2021SGR00801 Generalitat de Catalunya
- 1R01AG056850-01A1 CIBERNED Program 1, National Institutes of Health (NIH) grants
- 3RF1AG056850-01S1 CIBERNED Program 1, National Institutes of Health (NIH) grants
- AG056850 CIBERNED Program 1, National Institutes of Health (NIH) grants
- R01AG061566 CIBERNED Program 1, National Institutes of Health (NIH) grants
- R21AG056974 CIBERNED Program 1, National Institutes of Health (NIH) grants
- 888692 H2020 Marie Skłodowska-Curie Actions
- LCF/BQ/DR22/11950012 'la Caixa' Foundation
- PRE2021-099689 Ministerio de Ciencia e Innovación
- CEX2021-001159-M María de Maeztu Unit of Excellence (Institute of Neurosciences, University of Barcelona), Ministry of Science and Innovation
Collapse
Affiliation(s)
- Jèssica Pardo
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Victor Montal
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Life Sciences, Barcelona Supercomputing Center, Barcelona, Spain
| | - Anna Campabadal
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Neurology Service, Consorci Corporació Sanitària Parc Taulí de Sabadell, Barcelona, Spain
| | - Javier Oltra
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Carme Uribe
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | - Ignacio Roura
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Núria Bargalló
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Imaging Diagnostic Center (CDI), Hospital Clínic Universitari de Barcelona, Barcelona, Spain
| | - Maria J Martí
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Barcelona, Spain
- Parkinson's Disease and Movement Disorders Unit, Hospital Clínic Universitari de Barcelona, UBNeuro Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Yaroslau Compta
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Barcelona, Spain
- Parkinson's Disease and Movement Disorders Unit, Hospital Clínic Universitari de Barcelona, UBNeuro Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Alex Iranzo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Barcelona, Spain
- Sleep Disorders Center, Neurology Service, Hospital Clínic Universitari de Barcelona, University of Barcelona, Barcelona, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Barcelona, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Carme Junqué
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Barcelona, Spain
| | - Bàrbara Segura
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Barcelona, Spain
| |
Collapse
|
10
|
Cai Y, Shi D, Lan G, Chen L, Jiang Y, Zhou L, Guo T. Association of β-Amyloid, Microglial Activation, Cortical Thickness, and Metabolism in Older Adults Without Dementia. Neurology 2024; 102:e209205. [PMID: 38489560 DOI: 10.1212/wnl.0000000000209205] [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: 11/06/2023] [Accepted: 12/13/2023] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Plasma β-amyloid42 (Aβ42)/Aβ40 levels have shown promise in identifying Aβ-PET positive individuals. This study explored the concordance and discordance of plasma Aβ42/Aβ40 positivity (Plasma±) with CSF Aβ42/Aβ40 positivity (CSF±) and Aβ-PET positivity (PET±) in older adults without dementia. Associations of Aβ deposition, cortical thickness, glucose metabolism, and microglial activation were also investigated. METHODS We selected participants without dementia who had concurrent plasma Aβ42/Aβ40 and Aβ-PET scans from the Alzheimer's Disease Neuroimaging Initiative cohort. Participants were categorized into Plasma±/PET± based on thresholds of composite 18F-florbetapir (FBP) standardized uptake value ratio (SUVR) ≥1.11 and plasma Aβ42/Aβ40 ≤0.1218. Aβ-PET-negative individuals were further divided into Plasma±/CSF± (CSF Aβ42/Aβ40 ≤0.138), and the concordance and discordance of Aβ42/Aβ40 in the plasma and CSF were investigated. Baseline and slopes of regional FBP SUVR were compared among Plasma±/PET± groups, and associations of regional FBP SUVR, FDG SUVR, cortical thickness, and CSF soluble Triggering Receptor Expressed on Myeloid Cell 2 (sTREM2) levels were analyzed. RESULTS One hundred eighty participants (mean age 72.7 years, 51.4% female, 96 cognitively unimpaired, and 84 with mild cognitive impairment) were included. We found that the proportion of Plasma+/PET- individuals was 6.14 times higher (odds ratio (OR) = 6.143, 95% confidence interval (CI) 2.740-16.185, p < 0.001) than that of Plasma-/PET+ individuals, and Plasma+/CSF- individuals showed 8.5 times larger percentage (OR = 8.5, 95% CI: 3.031-32.974, p < 0.001) than Plasma-/CSF+ individuals in Aβ-PET-negative individuals. Besides, Plasma+/PET- individuals exhibited faster (p < 0.05) Aβ accumulation predominantly in bilateral banks of superior temporal sulcus (BANKSSTS) and supramarginal, and superior parietal cortices compared with Plasma-/PET- individuals, despite no difference in baseline FBP SUVRs. In Plasma+/PET+ individuals, higher CSF sTREM2 levels correlated with slower BANKSSTS Aβ accumulation (standardized β (βstd) = -0.418, 95% CI -0.681 to -0.154, p = 0.002). Conversely, thicker cortical thickness and higher glucose metabolism in supramarginal and superior parietal cortices were associated with faster (p < 0.05) CSF sTREM2 increase in Plasma+/PET- individuals rather than in Plasma+/PET+ individuals. DISCUSSION These findings suggest that plasma Aβ42/Aβ40 abnormalities may predate CSF Aβ42/Aβ40 and Aβ-PET abnormalities. Higher sTREM2-related microglial activation is linked to thicker cortical thickness and higher metabolism in early amyloidosis stages but tends to mitigate Aβ accumulation primarily at relatively advanced stages.
Collapse
Affiliation(s)
- Yue Cai
- From the Institute of Biomedical Engineering (Y.C., G.L., L.C., T.G.), Shenzhen Bay Laboratory; Neurology Medicine Center (D.S., L.Z.), The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Department of Psychology (Y.J.), University of Texas at Austin; and Institute of Biomedical Engineering (T.G.), Peking University Shenzhen Graduate School, China
| | - Dai Shi
- From the Institute of Biomedical Engineering (Y.C., G.L., L.C., T.G.), Shenzhen Bay Laboratory; Neurology Medicine Center (D.S., L.Z.), The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Department of Psychology (Y.J.), University of Texas at Austin; and Institute of Biomedical Engineering (T.G.), Peking University Shenzhen Graduate School, China
| | - Guoyu Lan
- From the Institute of Biomedical Engineering (Y.C., G.L., L.C., T.G.), Shenzhen Bay Laboratory; Neurology Medicine Center (D.S., L.Z.), The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Department of Psychology (Y.J.), University of Texas at Austin; and Institute of Biomedical Engineering (T.G.), Peking University Shenzhen Graduate School, China
| | - Linting Chen
- From the Institute of Biomedical Engineering (Y.C., G.L., L.C., T.G.), Shenzhen Bay Laboratory; Neurology Medicine Center (D.S., L.Z.), The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Department of Psychology (Y.J.), University of Texas at Austin; and Institute of Biomedical Engineering (T.G.), Peking University Shenzhen Graduate School, China
| | - Yanni Jiang
- From the Institute of Biomedical Engineering (Y.C., G.L., L.C., T.G.), Shenzhen Bay Laboratory; Neurology Medicine Center (D.S., L.Z.), The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Department of Psychology (Y.J.), University of Texas at Austin; and Institute of Biomedical Engineering (T.G.), Peking University Shenzhen Graduate School, China
| | - Liemin Zhou
- From the Institute of Biomedical Engineering (Y.C., G.L., L.C., T.G.), Shenzhen Bay Laboratory; Neurology Medicine Center (D.S., L.Z.), The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Department of Psychology (Y.J.), University of Texas at Austin; and Institute of Biomedical Engineering (T.G.), Peking University Shenzhen Graduate School, China
| | - Tengfei Guo
- From the Institute of Biomedical Engineering (Y.C., G.L., L.C., T.G.), Shenzhen Bay Laboratory; Neurology Medicine Center (D.S., L.Z.), The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Department of Psychology (Y.J.), University of Texas at Austin; and Institute of Biomedical Engineering (T.G.), Peking University Shenzhen Graduate School, China
| |
Collapse
|
11
|
Rodriguez-Vieitez E, Vannini P, Montal V, Graff C. Cortical microstructural imaging from diffusion MRI: towards sensitive biomarkers for clinical trials. Brain 2024; 147:746-748. [PMID: 38408356 DOI: 10.1093/brain/awae054] [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: 02/13/2024] [Accepted: 02/13/2024] [Indexed: 02/28/2024] Open
Abstract
This scientific commentary refers to ‘Diffusion MRI tracks cortical microstructural changes during the early stages of Alzheimer’s disease’ by Spotorno et al. (https://doi.org/10.1093/brain/awad428).
Collapse
Affiliation(s)
- Elena Rodriguez-Vieitez
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Patrizia Vannini
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Caroline Graff
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
12
|
Mak E, Dounavi ME, Operto G, Ziukelis ET, Jones PS, Low A, Swann P, Newton C, Muniz Terrera G, Malhotra P, Koychev I, Falcon C, Mackay C, Lawlor B, Naci L, Wells K, Ritchie C, Ritchie K, Su L, Gispert JD, O’Brien JT. APOE ɛ4 exacerbates age-dependent deficits in cortical microstructure. Brain Commun 2024; 6:fcad351. [PMID: 38384997 PMCID: PMC10881196 DOI: 10.1093/braincomms/fcad351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/20/2023] [Accepted: 02/09/2024] [Indexed: 02/23/2024] Open
Abstract
The apolipoprotein E ɛ4 allele is the primary genetic risk factor for the sporadic type of Alzheimer's disease. However, the mechanisms by which apolipoprotein E ɛ4 are associated with neurodegeneration are still poorly understood. We applied the Neurite Orientation Dispersion Model to characterize the effects of apolipoprotein ɛ4 and its interactions with age and education on cortical microstructure in cognitively normal individuals. Data from 1954 participants were included from the PREVENT-Dementia and ALFA (ALzheimer and FAmilies) studies (mean age = 57, 1197 non-carriers and 757 apolipoprotein E ɛ4 carriers). Structural MRI datasets were processed with FreeSurfer v7.2. The Microstructure Diffusion Toolbox was used to derive Orientation Dispersion Index maps from diffusion MRI datasets. Primary analyses were focused on (i) the main effects of apolipoprotein E ɛ4, and (ii) the interactions of apolipoprotein E ɛ4 with age and education on lobar and vertex-wise Orientation Dispersion Index and implemented using Permutation Analysis of Linear Models. There were apolipoprotein E ɛ4 × age interactions in the temporo-parietal and frontal lobes, indicating steeper age-dependent Orientation Dispersion Index changes in apolipoprotein E ɛ4 carriers. Steeper age-related Orientation Dispersion Index declines were observed among apolipoprotein E ɛ4 carriers with lower years of education. We demonstrated that apolipoprotein E ɛ4 worsened age-related Orientation Dispersion Index decreases in brain regions typically associated with atrophy patterns of Alzheimer's disease. This finding also suggests that apolipoprotein E ɛ4 may hasten the onset age of dementia by accelerating age-dependent reductions in cortical Orientation Dispersion Index.
Collapse
Affiliation(s)
- Elijah Mak
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Maria-Eleni Dounavi
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Grégory Operto
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona 08005, Spain
| | - Elina T Ziukelis
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Peter Simon Jones
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Audrey Low
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Peter Swann
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Coco Newton
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | | | - Paresh Malhotra
- Department of Brain Sciences, Imperial College, London W12 0NN, UK
| | - Ivan Koychev
- Department of Psychiatry, Oxford University, Oxford OX3 7JX, UK
| | - Carles Falcon
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona 08005, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona 08003, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
| | - Clare Mackay
- Department of Psychiatry, Oxford University, Oxford OX3 7JX, UK
| | - Brian Lawlor
- Institute of Neuroscience, Trinity College Dublin, University of Dublin, Dublin D02 PX31, Ireland
| | - Lorina Naci
- Institute of Neuroscience, Trinity College Dublin, University of Dublin, Dublin D02 PX31, Ireland
| | - Katie Wells
- Centre for Dementia Prevention, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Craig Ritchie
- Centre for Dementia Prevention, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Karen Ritchie
- Institut National de la Santé et de la Recherche Médicale, U1061 Neuropsychiatrie, Montpellier 34093, France
- Faculty of Medicine, University of Montpellier, Montpellier 34093, France
| | - Li Su
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona 08005, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona 08003, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
| | - John T O’Brien
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
13
|
Silva-Rudberg JA, Salardini E, O'Dell RS, Chen MK, Ra J, Georgelos JK, Morehouse MR, Melino KP, Varma P, Toyonaga T, Nabulsi NB, Huang Y, Carson RE, van Dyck CH, Mecca AP. Assessment of Gray Matter Microstructure and Synaptic Density in Alzheimer's Disease: A Multimodal Imaging Study With DTI and SV2A PET. Am J Geriatr Psychiatry 2024; 32:17-28. [PMID: 37673749 PMCID: PMC10840732 DOI: 10.1016/j.jagp.2023.08.002] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/19/2023] [Accepted: 08/05/2023] [Indexed: 09/08/2023]
Abstract
OBJECTIVE Multimodal imaging techniques have furthered our understanding of how different aspects of Alzheimer's disease (AD) pathology relate to one another. Diffusion tensor imaging (DTI) measures such as mean diffusivity (MD) may be a surrogate measure of the changes in gray matter structure associated with AD. Positron emission tomography (PET) imaging of synaptic vesicle glycoprotein 2A (SV2A) has been used to quantify synaptic loss, which is the major pathological correlate of cognitive impairment in AD. In this study, we investigated the relationship between gray matter microstructure and synaptic density. METHODS DTI was used to measure MD and [11C]UCB-J PET to measure synaptic density in 33 amyloid-positive participants with AD and 17 amyloid-negative cognitively normal (CN) participants aged 50-83. Univariate regression analyses were used to assess the association between synaptic density and MD in both the AD and CN groups. RESULTS Hippocampal MD was inversely associated with hippocampal synaptic density in participants with AD (r = -0.55, p <0.001, df = 31) but not CN (r = 0.13, p = 0.62, df = 15). Exploratory analyses across other regions known to be affected in AD suggested widespread inverse associations between synaptic density and MD in the AD group. CONCLUSION In the setting of AD, an increase in gray matter MD is inversely associated with synaptic density. These co-occurring changes may suggest a link between synaptic loss and gray matter microstructural changes in AD. Imaging studies of gray matter microstructure and synaptic density may allow important insights into AD-related neuropathology.
Collapse
Affiliation(s)
- Jason A Silva-Rudberg
- Alzheimer's Disease Research Unit (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Psychiatry (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT.
| | - Elaheh Salardini
- Alzheimer's Disease Research Unit (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Psychiatry (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT
| | - Ryan S O'Dell
- Alzheimer's Disease Research Unit (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Psychiatry (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT
| | - Ming-Kai Chen
- Department of Radiology and Biomedical Imaging (M-KC, PV, TT, NBN, YH, REC), Yale University School of Medicine, New Haven, CT
| | - Jocelyn Ra
- Alzheimer's Disease Research Unit (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Psychiatry (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT
| | - Jamie K Georgelos
- Alzheimer's Disease Research Unit (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Psychiatry (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT
| | - Mackenzie R Morehouse
- Alzheimer's Disease Research Unit (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Psychiatry (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT
| | - Kaitlyn P Melino
- Alzheimer's Disease Research Unit (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Psychiatry (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT
| | - Pradeep Varma
- Department of Radiology and Biomedical Imaging (M-KC, PV, TT, NBN, YH, REC), Yale University School of Medicine, New Haven, CT
| | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging (M-KC, PV, TT, NBN, YH, REC), Yale University School of Medicine, New Haven, CT
| | - Nabeel B Nabulsi
- Department of Radiology and Biomedical Imaging (M-KC, PV, TT, NBN, YH, REC), Yale University School of Medicine, New Haven, CT
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging (M-KC, PV, TT, NBN, YH, REC), Yale University School of Medicine, New Haven, CT
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging (M-KC, PV, TT, NBN, YH, REC), Yale University School of Medicine, New Haven, CT
| | - Christopher H van Dyck
- Alzheimer's Disease Research Unit (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Psychiatry (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Neuroscience (CHvD), Yale University School of Medicine, New Haven, CT; Department of Neurology (CHvD), Yale University School of Medicine, New Haven, CT
| | - Adam P Mecca
- Alzheimer's Disease Research Unit (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Psychiatry (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT.
| |
Collapse
|
14
|
Zammit MD, Betthauser TJ, McVea AK, Laymon CM, Tudorascu DL, Johnson SC, Hartley SL, Converse AK, Minhas DS, Zaman SH, Ances BM, Stone CK, Mathis CA, Cohen AD, Klunk WE, Handen BL, Christian BT. Characterizing the emergence of amyloid and tau burden in Down syndrome. Alzheimers Dement 2024; 20:388-398. [PMID: 37641577 PMCID: PMC10843570 DOI: 10.1002/alz.13444] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/14/2023] [Accepted: 07/23/2023] [Indexed: 08/31/2023]
Abstract
INTRODUCTION Almost all individuals with Down syndrome (DS) will develop neuropathological features of Alzheimer's disease (AD). Understanding AD biomarker trajectories is necessary for DS-specific clinical interventions and interpretation of drug-related changes in the disease trajectory. METHODS A total of 177 adults with DS from the Alzheimer's Biomarker Consortium-Down Syndrome (ABC-DS) underwent positron emission tomography (PET) and MR imaging. Amyloid-beta (Aβ) trajectories were modeled to provide individual-level estimates of Aβ-positive (A+) chronicity, which were compared against longitudinal tau change. RESULTS Elevated tau was observed in all NFT regions following A+ and longitudinal tau increased with respect to A+ chronicity. Tau increases in NFT regions I-III was observed 0-2.5 years following A+. Nearly all A+ individuals had tau increases in the medial temporal lobe. DISCUSSION These findings highlight the rapid accumulation of amyloid and early onset of tau relative to amyloid in DS and provide a strategy for temporally characterizing AD neuropathology progression that is specific to the DS population and independent of chronological age. HIGHLIGHTS Longitudinal amyloid trajectories reveal rapid Aβ accumulation in Down syndrome NFT stage tau was strongly associated with A+ chronicity Early longitudinal tau increases were observed 2.5-5 years after reaching A.
Collapse
Affiliation(s)
| | - Tobey J. Betthauser
- University of Wisconsin‐Madison Alzheimer's Disease Research CenterMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Andrew K. McVea
- University of Wisconsin‐Madison Waisman CenterMadisonWisconsinUSA
| | - Charles M. Laymon
- Department of RadiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Dana L. Tudorascu
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Sterling C. Johnson
- University of Wisconsin‐Madison Alzheimer's Disease Research CenterMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Sigan L. Hartley
- University of Wisconsin‐Madison Waisman CenterMadisonWisconsinUSA
| | | | - Davneet S. Minhas
- Department of RadiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Shahid H. Zaman
- Cambridge Intellectual Disability Research GroupUniversity of CambridgeCambridgeUK
| | - Beau M. Ances
- Department of NeurologyWashington University in St. LouisSt. LouisMissouriUSA
| | - Charles K. Stone
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Chester A. Mathis
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Annie D. Cohen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - William E. Klunk
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Benjamin L. Handen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Bradley T. Christian
- University of Wisconsin‐Madison Waisman CenterMadisonWisconsinUSA
- Department of Medical PhysicsUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | | |
Collapse
|
15
|
Gagliardi G, Rodriguez-Vieitez E, Montal V, Sepulcre J, Diez I, Lois C, Hanseeuw B, Schultz AP, Properzi MJ, Papp KV, Marshall GA, Fortea J, Johnson KA, Sperling RA, Vannini P. Cortical microstructural changes predict tau accumulation and episodic memory decline in older adults harboring amyloid. COMMUNICATIONS MEDICINE 2023; 3:106. [PMID: 37528163 PMCID: PMC10394044 DOI: 10.1038/s43856-023-00324-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/19/2023] [Indexed: 08/03/2023] Open
Abstract
INTRODUCTION Non-invasive diffusion-weighted imaging (DWI) to assess brain microstructural changes via cortical mean diffusivity (cMD) has been shown to be cross-sectionally associated with tau in cognitively normal older adults, suggesting that it might be an early marker of neuronal injury. Here, we investigated how regional cortical microstructural changes measured by cMD are related to the longitudinal accumulation of regional tau as well as to episodic memory decline in cognitively normal individuals harboring amyloid pathology. METHODS 122 cognitively normal participants from the Harvard Aging Brain Study underwent DWI, T1w-MRI, amyloid and tau PET imaging, and Logical Memory Delayed Recall (LMDR) assessments. We assessed whether the interaction of baseline amyloid status and cMD (in entorhinal and inferior-temporal cortices) was associated with longitudinal regional tau accumulation and with longitudinal LMDR using separate linear mixed-effects models. RESULTS We find a significant interaction effect of the amyloid status and baseline cMD in predicting longitudinal tau in the entorhinal cortex (p = 0.044) but not the inferior temporal lobe, such that greater baseline cMD values predicts the accumulation of entorhinal tau in amyloid-positive participants. Moreover, we find a significant interaction effect of the amyloid status and baseline cMD in the entorhinal cortex (but not inferior temporal cMD) in predicting longitudinal LMDR (p < 0.001), such that baseline entorhinal cMD predicts the episodic memory decline in amyloid-positive participants. CONCLUSIONS The combination of amyloidosis and elevated cMD in the entorhinal cortex may help identify individuals at short-term risk of tau accumulation and Alzheimer's Disease-related episodic memory decline, suggesting utility in clinical trials.
Collapse
Affiliation(s)
- Geoffroy Gagliardi
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, 02129, USA
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Elena Rodriguez-Vieitez
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, 02129, USA
- Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, Stockholm, 14152, Sweden
| | - Victor Montal
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, 08041, Spain
- Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, 28031, Spain
| | - Jorge Sepulcre
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Gordon Center for Medical Imaging, Boston, MA, 02114, USA
| | - Ibai Diez
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Gordon Center for Medical Imaging, Boston, MA, 02114, USA
| | - Cristina Lois
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Gordon Center for Medical Imaging, Boston, MA, 02114, USA
| | - Bernard Hanseeuw
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Gordon Center for Medical Imaging, Boston, MA, 02114, USA
- Saint Luc University Hospital, Université Catholique de Louvain, Brussels, 1200, Belgium
| | - Aaron P Schultz
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, 02129, USA
| | - Michael J Properzi
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Kathryn V Papp
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, 02129, USA
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Gad A Marshall
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, 08041, Spain
- Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, 28031, Spain
| | - Keith A Johnson
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Gordon Center for Medical Imaging, Boston, MA, 02114, USA
| | - Reisa A Sperling
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, 02129, USA
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Patrizia Vannini
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, 02129, USA.
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
16
|
Swaddiwudhipong N, Whiteside DJ, Hezemans FH, Street D, Rowe JB, Rittman T. Pre-diagnostic cognitive and functional impairment in multiple sporadic neurodegenerative diseases. Alzheimers Dement 2023; 19:1752-1763. [PMID: 36223793 DOI: 10.1002/alz.12802] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
INTRODUCTION The pathophysiological processes of neurodegenerative diseases begin years before diagnosis. However, pre-diagnostic changes in cognition and physical function are poorly understood, especially in sporadic neurodegenerative disease. METHODS UK Biobank data were extracted. Cognitive and functional measures in individuals who subsequently developed Alzheimer's disease (AD), Parkinson disease, frontotemporal dementia, progressive supranuclear palsy, dementia with Lewy bodies, or multiple system atrophy were compared against individuals without neurodegenerative diagnoses. The same measures were regressed against time to diagnosis, after adjusting for the effects of age. RESULTS There was evidence for pre-diagnostic cognitive impairment and decline with time, particularly in AD. Pre-diagnostic functional impairment and decline were observed in multiple diseases. DISCUSSION The scale and longitudinal follow-up of UK Biobank participants provides evidence for cognitive and functional decline years before symptoms become obvious in multiple neurodegenerative diseases. Identifying pre-diagnostic functional and cognitive changes could improve selection for preventive and early disease-modifying treatment trials.
Collapse
Affiliation(s)
- Nol Swaddiwudhipong
- Department of Clinical Neurosciences, Cambridge, UK
- Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
| | - David J Whiteside
- Department of Clinical Neurosciences, Cambridge, UK
- Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
| | - Frank H Hezemans
- Department of Clinical Neurosciences, Cambridge, UK
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | | | - James B Rowe
- Department of Clinical Neurosciences, Cambridge, UK
- Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Timothy Rittman
- Department of Clinical Neurosciences, Cambridge, UK
- Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
| |
Collapse
|
17
|
Ding W, Ren P, Yi L, Si Y, Yang F, Li Z, Bao H, Yan S, Zhang X, Li S, Liang X, Yao L. Association of cortical and subcortical microstructure with disease severity: impact on cognitive decline and language impairments in frontotemporal lobar degeneration. Alzheimers Res Ther 2023; 15:58. [PMID: 36941645 PMCID: PMC10029187 DOI: 10.1186/s13195-023-01208-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 03/13/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND Cortical and subcortical microstructural modifications are critical to understanding the pathogenic changes in frontotemporal lobar degeneration (FTLD) subtypes. In this study, we investigated cortical and subcortical microstructure underlying cognitive and language impairments across behavioral variant of frontotemporal dementia (bvFTD), semantic variant of primary progressive aphasia (svPPA), and nonfluent variant of primary progressive aphasia (nfvPPA) subtypes. METHODS The current study characterized 170 individuals with 3 T MRI structural and diffusion-weighted imaging sequences as portion of the Frontotemporal Lobar Degeneration Neuroimaging Initiative study: 41 bvFTD, 35 nfvPPA, 34 svPPA, and 60 age-matched cognitively unimpaired controls. To determine the severity of the disease, clinical dementia rating plus national Alzheimer's coordinating center behavior and language domains sum of boxes scores were used; other clinical measures, including the Boston naming test and verbal fluency test, were also evaluated. We computed surface-based cortical thickness and cortical and subcortical microstructural metrics using tract-based spatial statistics and explored their relationships with clinical and cognitive assessments. RESULTS Compared with controls, those with FTLD showed substantial cortical mean diffusivity alterations extending outside the regions with cortical thinning. Tract-based spatial statistics revealed that anomalies in subcortical white matter diffusion were widely distributed across the frontotemporal and parietal areas. Patients with bvFTD, nfvPPA, and svPPA exhibited distinct patterns of cortical and subcortical microstructural abnormalities, which appeared to correlate with disease severity, and separate dimensions of language functions. CONCLUSIONS Our findings imply that cortical and subcortical microstructures may serve as sensitive biomarkers for the investigation of neurodegeneration-associated microstructural alterations in FTLD subtypes. Flowchart of the study design (see materials and methods for detailed description).
Collapse
Affiliation(s)
- Wencai Ding
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Peng Ren
- Laboratory for Space Environment and Physical Science, Harbin Institute of Technology, Harbin, 150001, China
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Liye Yi
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Yao Si
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Fan Yang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Zhipeng Li
- Laboratory for Space Environment and Physical Science, Harbin Institute of Technology, Harbin, 150001, China
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Hongbo Bao
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Shi Yan
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xinyu Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Siyang Li
- Laboratory for Space Environment and Physical Science, Harbin Institute of Technology, Harbin, 150001, China
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Xia Liang
- Laboratory for Space Environment and Physical Science, Harbin Institute of Technology, Harbin, 150001, China.
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China.
| | - Lifen Yao
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
18
|
Spatial and Temporal Protein Modules Signatures Associated with Alzheimer Disease in 3xTg-AD Mice Are Restored by Early Ubiquinol Supplementation. Antioxidants (Basel) 2023; 12:antiox12030747. [PMID: 36978996 PMCID: PMC10044705 DOI: 10.3390/antiox12030747] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/22/2023] Open
Abstract
Despite its robust proteopathic nature, the spatiotemporal signature of disrupted protein modules in sporadic Alzheimer’s disease (AD) brains remains poorly understood. This considered oxidative stress contributes to AD progression and early intervention with coenzyme Q10 or its reduced form, ubiquinol, delays the progression of the disease. Using MALDI–MSI and functional bioinformatic analysis, we have developed a protocol to express how deregulated protein modules arise from hippocampus and cortex in the AD mice model 3xTG-AD in an age-dependent manner. This strategy allowed us to identify which modules can be efficiently restored to a non-pathological condition by early intervention with ubiquinol. Indeed, an early deregulation of proteostasis-related protein modules, oxidative stress and metabolism has been observed in the hippocampus of 6-month mice (early AD) and the mirrored in cortical regions of 12-month mice (middle/late AD). This observation has been validated by IHC using mouse and human brain sections, suggesting that these protein modules are also affected in humans. The emergence of disrupted protein modules with AD signature can be prevented by early dietary intervention with ubiquinol in the 3xTG-AD mice model.
Collapse
|
19
|
Zetterberg H, Teunissen C, van Swieten J, Kuhle J, Boxer A, Rohrer JD, Mitic L, Nicholson AM, Pearlman R, McCaughey SM, Tatton N. The role of neurofilament light in genetic frontotemporal lobar degeneration. Brain Commun 2023; 5:fcac310. [PMID: 36694576 PMCID: PMC9866262 DOI: 10.1093/braincomms/fcac310] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 09/26/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
Genetic frontotemporal lobar degeneration caused by autosomal dominant gene mutations provides an opportunity for targeted drug development in a highly complex and clinically heterogeneous dementia. These neurodegenerative disorders can affect adults in their middle years, progress quickly relative to other dementias, are uniformly fatal and have no approved disease-modifying treatments. Frontotemporal dementia, caused by mutations in the GRN gene which encodes the protein progranulin, is an active area of interventional drug trials that are testing multiple strategies to restore progranulin protein deficiency. These and other trials are also examining neurofilament light as a potential biomarker of disease activity and disease progression and as a therapeutic endpoint based on the assumption that cerebrospinal fluid and blood neurofilament light levels are a surrogate for neuroaxonal damage. Reports from genetic frontotemporal dementia longitudinal studies indicate that elevated concentrations of blood neurofilament light reflect disease severity and are associated with faster brain atrophy. To better inform patient stratification and treatment response in current and upcoming clinical trials, a more nuanced interpretation of neurofilament light as a biomarker of neurodegeneration is now required, one that takes into account its relationship to other pathophysiological and topographic biomarkers of disease progression from early presymptomatic to later clinically symptomatic stages.
Collapse
Affiliation(s)
- Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden.,Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden.,Dementia Research Institute, University College London, London, UK.,DRI Fluid Biomarker Laboratory, Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Charlotte Teunissen
- Department of Clinical Chemistry, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - John van Swieten
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Jens Kuhle
- Department of Clinical Research, Department of Neurology, Department of Biomedicine, Multiple Sclerosis Centre, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Adam Boxer
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Jonathan D Rohrer
- Queen Square UCL Institute of Neurology, Dementia Research Centre, UK Dementia Research Institute, University College London, London, UK
| | - Laura Mitic
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, USA.,The Bluefield Project to Cure FTD, San Francisco, CA, USA
| | - Alexandra M Nicholson
- The Bluefield Project to Cure FTD, San Francisco, CA, USA.,Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | | | | | - Nadine Tatton
- Medical Affairs, Alector, Inc., South San Francisco, CA, USA
| |
Collapse
|
20
|
Buglio DS, Marton LT, Laurindo LF, Guiguer EL, Araújo AC, Buchaim RL, Goulart RDA, Rubira CJ, Barbalho SM. The Role of Resveratrol in Mild Cognitive Impairment and Alzheimer's Disease: A Systematic Review. J Med Food 2022; 25:797-806. [PMID: 35353606 DOI: 10.1089/jmf.2021.0084] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Advancing age is one of the risk factors for developing many diseases, including cancer, cardiovascular disorders, and neurodegenerative alterations, such as mild cognitive impairment (MCI), and Alzheimer's Disease (AD). Studies have indicated that supplementation with resveratrol (RSV) might improve cerebrovascular function and reduce the risk of developing dementia. Thus, the aim of this systematic review was to assess the effects of RSV on MCI and AD. MEDLINE-PubMed, Cochrane, and EMBASE were used to perform the search, and PRISMA guidelines were followed. Five studies met the eligible criteria; three with AD and two with MCI. In AD patients, the use of RSV reduces Aβ levels, improves brain volume, reduces the Mini-mental status score, and improves AD scores. In patients with MCI, this polyphenol prevents decline in Standard Volumes of Interest and increases the Resting-state Functional Connectivity score. RSV can activate the human silent information regulator 2/sirtuin 1 (Sirt-1) and can inhibit the cyclooxygenase-2 (COX-2), 5-lipoxygenase, and nuclear factor-κB, resulting in the reduction of the proinflammation pathways. It is also associated with the increase in the levels of interleukin (IL)-10 and reduction of interferon-γ and IL-17. Both anti-inflammatory and antioxidant effects can be related to preventing neurodegenerative diseases, doing maintenance, and enabling the recovery of these conditions directly related to inflammation and oxidative stress. We suggest that the use of RSV can bring beneficial effects to patients with MCI or AD.
Collapse
Affiliation(s)
- Daiene Santos Buglio
- Structural and Functional Interactions in Rehabilitation-UNIMAR, Marília, São Paulo, Brazil
| | - Ledyane Taynara Marton
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Elen Landgraf Guiguer
- Structural and Functional Interactions in Rehabilitation-UNIMAR, Marília, São Paulo, Brazil
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília, São Paulo, Brazil
- School of Food and Technology of Marilia (FATEC), Marilia, São Paulo, Brazil
| | - Adriano Cressoni Araújo
- Structural and Functional Interactions in Rehabilitation-UNIMAR, Marília, São Paulo, Brazil
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Rogério Leone Buchaim
- Department of Biological Sciences, Bauru School of Dentistry (FOB/USP), University of São Paulo, Bauru, São Paulo, Brazil
| | | | - Cláudio José Rubira
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Sandra M Barbalho
- Structural and Functional Interactions in Rehabilitation-UNIMAR, Marília, São Paulo, Brazil
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília, São Paulo, Brazil
- School of Food and Technology of Marilia (FATEC), Marilia, São Paulo, Brazil
| |
Collapse
|
21
|
Montal V, Diez I, Kim CM, Orwig W, Bueichekú E, Gutiérrez-Zúñiga R, Bejanin A, Pegueroles J, Dols-Icardo O, Vannini P, El-Fakhri G, Johnson KA, Sperling RA, Fortea J, Sepulcre J. Network Tau spreading is vulnerable to the expression gradients of APOE and glutamatergic-related genes. Sci Transl Med 2022; 14:eabn7273. [PMID: 35895837 PMCID: PMC9942690 DOI: 10.1126/scitranslmed.abn7273] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
A key hallmark of Alzheimer's disease (AD) pathology is the intracellular accumulation of tau protein in the form of neurofibrillary tangles across large-scale networks of the human brain cortex. Currently, it is still unclear how tau accumulates within specific cortical systems and whether in situ genetic traits play a role in this circuit-based propagation progression. In this study, using two independent cohorts of cognitively normal older participants, we reveal the brain network foundation of tau spreading and its association with using high-resolution transcriptomic genetic data. We observed that specific connectomic and genetic gradients exist along the tau spreading network. In particular, we identified 577 genes whose expression is associated with the spatial spreading of tau. Within this set of genes, APOE and glutamatergic synaptic genes, such as SLC1A2, play a central role. Thus, our study characterizes neurogenetic topological vulnerabilities in distinctive brain circuits of tau spreading and suggests that drug development strategies targeting the gradient expression of this set of genes should be explored to help reduce or prevent pathological tau accumulation.
Collapse
Affiliation(s)
- Victor Montal
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School; Boston, Massachusetts, USA.,Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autonoma de Barcelona; Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED); Madrid, Spain
| | - Ibai Diez
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School; Boston, Massachusetts, USA.,Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School; Charlestown, Massachusetts, USA
| | - Chan-Mi Kim
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School; Boston, Massachusetts, USA
| | - William Orwig
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School; Boston, Massachusetts, USA
| | - Elisenda Bueichekú
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School; Boston, Massachusetts, USA
| | - Raquel Gutiérrez-Zúñiga
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School; Boston, Massachusetts, USA
| | - Alexandre Bejanin
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autonoma de Barcelona; Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED); Madrid, Spain
| | - Jordi Pegueroles
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autonoma de Barcelona; Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED); Madrid, Spain
| | - Oriol Dols-Icardo
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autonoma de Barcelona; Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED); Madrid, Spain
| | - Patrizia Vannini
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School; Charlestown, Massachusetts, USA.,Center for Alzheimer research and treatment, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School; Boston, MA
| | - Georges El-Fakhri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School; Boston, Massachusetts, USA
| | - Keith A. Johnson
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School; Boston, Massachusetts, USA
| | - Reisa A. Sperling
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School; Charlestown, Massachusetts, USA
| | - Juan Fortea
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autonoma de Barcelona; Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED); Madrid, Spain
| | - Jorge Sepulcre
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School; Boston, Massachusetts, USA.,Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School; Charlestown, Massachusetts, USA.,Correspondence should be addressed to Jorge Sepulcre, 149 13th St, Office 5.209, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts; ; +1 617 726 2899
| |
Collapse
|
22
|
Padilla C, Montal V, Walpert MJ, Hong YT, Fryer TD, Coles JP, Aigbirhio FI, Hartley SL, Cohen AD, Tudorascu DL, Christian BT, Handen BL, Klunk WE, Holland AJ, Zaman SH. Cortical atrophy and amyloid and tau deposition in Down syndrome: A longitudinal study. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12288. [PMID: 35386472 PMCID: PMC8974205 DOI: 10.1002/dad2.12288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/30/2021] [Accepted: 01/10/2022] [Indexed: 11/17/2022]
Abstract
Introduction: The Down syndrome population has a high prevalence for dementia, often showing their first clinical symptoms in their 40s. Methods: In a longitudinal cohort, we investigate whether amyloid deposition at time point 1 (TP1) could predict cortical thickness change at time point 2 (TP2). The association between tau burden and cortical thickness was also examined at time point 3 (TP3). Results: Between TP1 and TP2 there was pronounced cortical thinning in temporo-parietal cortices and cortical thickening in the frontal cortex. Baseline amyloid burden was strongly associated to cortical thinning progression, especially in the temporo-parietal regions. At TP3, tau deposition negatively correlated with cortical atrophy in regions where tau usually accumulates at later Braak stages. Discussion: A higher amount of amyloid accumulation triggers a cascade of changes of disease-causing processes that eventually lead to dementia. As expected, we found that regions where tau usually accumulates were those also displaying high levels of cortical atrophy.
Collapse
Affiliation(s)
- Concepcion Padilla
- Cambridge Intellectual and Developmental Disabilities Research Group, Department of PsychiatryUniversity of CambridgeCambridgeUK
- Memory Unit and Biomedical Research Institute Sant Pau (IIB Sant Pau), Neurology DepartmentSanta Creu and Sant Pau HospitalBarcelonaSpain
- The Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED)MadridSpain
| | - Victor Montal
- Memory Unit and Biomedical Research Institute Sant Pau (IIB Sant Pau), Neurology DepartmentSanta Creu and Sant Pau HospitalBarcelonaSpain
- The Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED)MadridSpain
| | - Madeleine J. Walpert
- Cambridge Intellectual and Developmental Disabilities Research Group, Department of PsychiatryUniversity of CambridgeCambridgeUK
| | - Young T. Hong
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, Cambridge Biomedical CampusUniversity of CambridgeCambridgeUK
| | - Tim D. Fryer
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, Cambridge Biomedical CampusUniversity of CambridgeCambridgeUK
| | - Jonathan P. Coles
- Division of Anaesthesia, Department of Medicine, Cambridge Biomedical CampusUniversity of CambridgeCambridgeUK
| | - Franklin I. Aigbirhio
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, Cambridge Biomedical CampusUniversity of CambridgeCambridgeUK
| | - Sigan L. Hartley
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Ann D. Cohen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Dana L Tudorascu
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | | | - Benjamin L. Handen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - William E. Klunk
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Anthony J. Holland
- Cambridge Intellectual and Developmental Disabilities Research Group, Department of PsychiatryUniversity of CambridgeCambridgeUK
- Cambridgeshire and Peterborough NHS Foundation TrustFulbourn HospitalCambridgeUK
| | - Shahid H. Zaman
- Cambridge Intellectual and Developmental Disabilities Research Group, Department of PsychiatryUniversity of CambridgeCambridgeUK
- Cambridgeshire and Peterborough NHS Foundation TrustFulbourn HospitalCambridgeUK
| |
Collapse
|
23
|
Illán-Gala I, Montal V, Borrego-Écija S, Mandelli ML, Falgàs N, Welch AE, Pegueroles J, Santos-Santos M, Bejanin A, Alcolea D, Dols-Icardo O, Belbin O, Sánchez-Saudinós MB, Bargalló N, González-Ortiz S, Lladó A, Blesa R, Dickerson BC, Rosen HJ, Miller BL, Lleó A, Gorno-Tempini ML, Sánchez-Valle R, Fortea J. Cortical microstructure in primary progressive aphasia: a multicenter study. Alzheimers Res Ther 2022; 14:27. [PMID: 35139897 PMCID: PMC8830043 DOI: 10.1186/s13195-022-00974-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/02/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cortical mean diffusivity is a novel imaging metric sensitive to early changes in neurodegenerative syndromes. Higher cortical mean diffusivity values reflect microstructural disorganization and have been proposed as a sensitive biomarker that might antedate macroscopic cortical changes. We aimed to test the hypothesis that cortical mean diffusivity is more sensitive than cortical thickness to detect cortical changes in primary progressive aphasia (PPA). METHODS In this multicenter, case-control study, we recruited 120 patients with PPA (52 non-fluent, 31 semantic, and 32 logopenic variants; and 5 GRN-related PPA) as well as 89 controls from three centers. The 3-Tesla MRI protocol included structural and diffusion-weighted sequences. Disease severity was assessed with the Clinical Dementia Rating scale. Cortical thickness and cortical mean diffusivity were computed using a surface-based approach. RESULTS The comparison between each PPA variant and controls revealed cortical mean diffusivity increases and cortical thinning in overlapping regions, reflecting the canonical loci of neurodegeneration of each variant. Importantly, cortical mean diffusivity increases also expanded to other PPA-related areas and correlated with disease severity in all PPA groups. Cortical mean diffusivity was also increased in patients with very mild PPA when only minimal cortical thinning was observed and showed a good correlation with measures of disease severity. CONCLUSIONS Cortical mean diffusivity shows promise as a sensitive biomarker for the study of the neurodegeneration-related microstructural changes in PPA.
Collapse
Affiliation(s)
- Ignacio Illán-Gala
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain.
- Atlantic Fellow for Equity in Brain Health at the University of California San Francisco, San Francisco, CA, 94115, USA.
| | - Victor Montal
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Sergi Borrego-Écija
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Alzheimer's Disease and Other Cognitive Disorders Unit, Service of Neurology, Hospital Clínic de Barcelona, Institut d'Investigació Biomèdica August Pi i Sunyer, University of Barcelona, 08036, Barcelona, Spain
| | - Maria Luisa Mandelli
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, 94115, USA
| | - Neus Falgàs
- Atlantic Fellow for Equity in Brain Health at the University of California San Francisco, San Francisco, CA, 94115, USA
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, 94115, USA
| | - Ariane E Welch
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, 94115, USA
| | - Jordi Pegueroles
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Miguel Santos-Santos
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Alexandre Bejanin
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Daniel Alcolea
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Oriol Dols-Icardo
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Olivia Belbin
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Mª Belén Sánchez-Saudinós
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain
| | - Nuria Bargalló
- Radiology Department, Hospital Clinic Barcelona and Magnetic Resonance Image Core facility, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Albert Lladó
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Alzheimer's Disease and Other Cognitive Disorders Unit, Service of Neurology, Hospital Clínic de Barcelona, Institut d'Investigació Biomèdica August Pi i Sunyer, University of Barcelona, 08036, Barcelona, Spain
| | - Rafael Blesa
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Bradford C Dickerson
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA
| | - Howard J Rosen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, 94115, USA
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, 94115, USA
| | - Alberto Lleó
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Maria Luisa Gorno-Tempini
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, 94115, USA
| | - Raquel Sánchez-Valle
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Alzheimer's Disease and Other Cognitive Disorders Unit, Service of Neurology, Hospital Clínic de Barcelona, Institut d'Investigació Biomèdica August Pi i Sunyer, University of Barcelona, 08036, Barcelona, Spain
| | - Juan Fortea
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain.
- Barcelona Down Medical Center. Fundació Catalana de Síndrome de Down, Barcelona, Spain.
| |
Collapse
|
24
|
Reyes-Leiva D, Dols-Icardo O, Sirisi S, Cortés-Vicente E, Turon-Sans J, de Luna N, Blesa R, Belbin O, Montal V, Alcolea D, Fortea J, Lleó A, Rojas-García R, Illán-Gala I. Pathophysiological Underpinnings of Extra-Motor Neurodegeneration in Amyotrophic Lateral Sclerosis: New Insights From Biomarker Studies. Front Neurol 2022; 12:750543. [PMID: 35115992 PMCID: PMC8804092 DOI: 10.3389/fneur.2021.750543] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 12/09/2021] [Indexed: 11/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) lie at opposing ends of a clinical, genetic, and neuropathological continuum. In the last decade, it has become clear that cognitive and behavioral changes in patients with ALS are more frequent than previously recognized. Significantly, these non-motor features can impact the diagnosis, prognosis, and management of ALS. Partially overlapping neuropathological staging systems have been proposed to describe the distribution of TAR DNA-binding protein 43 (TDP-43) aggregates outside the corticospinal tract. However, the relationship between TDP-43 inclusions and neurodegeneration is not absolute and other pathophysiological processes, such as neuroinflammation (with a prominent role of microglia), cortical hyperexcitability, and synaptic dysfunction also play a central role in ALS pathophysiology. In the last decade, imaging and biofluid biomarker studies have revealed important insights into the pathophysiological underpinnings of extra-motor neurodegeneration in the ALS-FTLD continuum. In this review, we first summarize the clinical and pathophysiological correlates of extra-motor neurodegeneration in ALS. Next, we discuss the diagnostic and prognostic value of biomarkers in ALS and their potential to characterize extra-motor neurodegeneration. Finally, we debate about how biomarkers could improve the diagnosis and classification of ALS. Emerging imaging biomarkers of extra-motor neurodegeneration that enable the monitoring of disease progression are particularly promising. In addition, a growing arsenal of biofluid biomarkers linked to neurodegeneration and neuroinflammation are improving the diagnostic accuracy and identification of patients with a faster progression rate. The development and validation of biomarkers that detect the pathological aggregates of TDP-43 in vivo are notably expected to further elucidate the pathophysiological underpinnings of extra-motor neurodegeneration in ALS. Novel biomarkers tracking the different aspects of ALS pathophysiology are paving the way to precision medicine approaches in the ALS-FTLD continuum. These are essential steps to improve the diagnosis and staging of ALS and the design of clinical trials testing novel disease-modifying treatments.
Collapse
Affiliation(s)
- David Reyes-Leiva
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Oriol Dols-Icardo
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Sonia Sirisi
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Elena Cortés-Vicente
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Janina Turon-Sans
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Noemi de Luna
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Rafael Blesa
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Olivia Belbin
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Victor Montal
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Ricard Rojas-García
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Ignacio Illán-Gala
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
- *Correspondence: Ignacio Illán-Gala
| |
Collapse
|
25
|
Increased homocysteine levels correlate with cortical structural damage in Parkinson's disease. J Neurol Sci 2022; 434:120148. [DOI: 10.1016/j.jns.2022.120148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/07/2021] [Accepted: 01/05/2022] [Indexed: 12/12/2022]
|
26
|
Chen C, Lu J, Peng W, Mak MS, Yang Y, Zhu Z, Wang S, Hou J, Zhou X, Xin W, Hu Y, Tsim KWK, Han Y, Liu Q, Pi R. Acrolein, an endogenous aldehyde induces Alzheimer's disease-like pathologies in mice: A new sporadic AD animal model. Pharmacol Res 2022; 175:106003. [PMID: 34838693 DOI: 10.1016/j.phrs.2021.106003] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/01/2021] [Accepted: 11/22/2021] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease that mainly affects elderly people. However, the translational research of AD is frustrating, suggesting that the development of new AD animal models is crucial. By gavage administration of acrolein, we constructed a simple sporadic AD animal model which showed classic pathologies of AD in 1 month. The AD-like phenotypes and pathological changes were as followed. 1) olfactory dysfunctions, cognitive impairments and psychological symptoms in C57BL/6 mice; 2) increased levels of Aβ1-42 and Tau phosphorylation (S396/T231) in cortex and hippocampus; 3) astrocytes and microglia proliferation; 4) reduced levels of postsynaptic density 95(PSD95) and Synapsin1, as well as the density of dendritic spines in the CA1 and DG neurons of the hippocampus; 5) high-frequency stimulation failed to induce the long-term potentiation (LTP) in the hippocampus after exposure to acrolein for 4 weeks; 6) decreased blood oxygen level-dependent (BOLD) signal in the olfactory bulb and induced high T2 signals in the hippocampus, which matched to the clinical observation in the brain of AD patients, and 7) activated RhoA/ROCK2/ p-cofilin-associated pathway in hippocampus of acrolein-treated mice, which may be the causes of synaptic damage and neuroinflammation in acrolein mice model. Taken together, the acrolein-induced sporadic AD mouse model closely reflects the pathological features of AD, which will be useful for the research on the mechanism of AD onset and the development of anti-AD drugs.
Collapse
Affiliation(s)
- Chen Chen
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Junfeng Lu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Weijia Peng
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Marvin Sh Mak
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yang Yang
- Department of Pharmacology, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China; Neurobiology Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China
| | - Zeyu Zhu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Shuyi Wang
- Department of Pharmacology, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China; Neurobiology Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China
| | - Jiawei Hou
- Neurobiology Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China
| | - Xin Zhou
- Zhongshan school of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Wenjun Xin
- Zhongshan school of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yafang Hu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510014, China
| | - Karl Wah Keung Tsim
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yifan Han
- Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hong Kong, China
| | - Qinyu Liu
- The seventh affiliated hospital, Sun Yat-Sen University, Shenzhen 518107, China.
| | - Rongbiao Pi
- Department of Pharmacology, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China; International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangzhou, Guangzhou 510006, China; Neurobiology Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China.
| |
Collapse
|
27
|
Fox-Fuller JT, Torrico-Teave H, d'Oleire Uquillas F, Chen K, Su Y, Chen Y, Brickhouse M, Sanchez JS, Aguero C, Jacobs HIL, Hampton O, Guzmán-Vélez E, Vila-Castelar C, Aguirre-Acevedo DC, Baena A, Artola A, Martinez J, Pluim CF, Alvarez S, Ochoa-Escudero M, Reiman EM, Sperling RA, Lopera F, Johnson KA, Dickerson BC, Quiroz YT. Cortical thickness across the lifespan in a Colombian cohort with autosomal-dominant Alzheimer's disease: A cross-sectional study. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12233. [PMID: 34541287 PMCID: PMC8438687 DOI: 10.1002/dad2.12233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/22/2021] [Accepted: 06/28/2021] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Cortical thinning is a marker of neurodegeneration in Alzheimer's disease (AD). We investigated the age-related trajectory of cortical thickness across the lifespan (9-59 years) in a Colombian kindred with autosomal dominant AD (ADAD). METHODS Two hundred eleven participants (105 presenilin-1 [PSEN1] E280A mutation carriers, 16 with cognitive impairment; 106 non-carriers) underwent magnetic resonance imaging. A piecewise linear regression identified change-points in the age-related trajectory of cortical thickness in carriers and non-carriers. RESULTS Unimpaired carriers exhibited elevated cortical thickness compared to non-carriers, and thickness more negatively correlated with age and cognition in carriers relative to non-carriers. We found increased cortical thickness in child carriers, after which thickness steadied compared to non-carriers prior to a rapid reduction in the decade leading up to the expected age at cognitive impairment in carriers. DISCUSSION Findings suggest that cortical thickness may fluctuate across the ADAD lifespan, from early-life increased thickness to atrophy proximal to clinical onset.
Collapse
Affiliation(s)
- Joshua T Fox-Fuller
- Department of Psychological and Brain Sciences Boston University Boston Massachusetts USA
- Department of Psychiatry Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
| | - Heirangi Torrico-Teave
- Department of Psychiatry Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
| | - Federico d'Oleire Uquillas
- Department of Neurology Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
- Princeton Neuroscience Institute Princeton University Princeton New Jersey USA
| | - Kewei Chen
- Banner Alzheimer's Institute Phoenix Arizona USA
| | - Yi Su
- Banner Alzheimer's Institute Phoenix Arizona USA
| | - Yinghua Chen
- Banner Alzheimer's Institute Phoenix Arizona USA
| | - Michael Brickhouse
- Department of Neurology Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
| | - Justin S Sanchez
- Department of Neurology Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
| | - Cinthya Aguero
- MassGeneral Institute for Neurodegenerative Disease Charlestown Massachusetts USA
| | - Heidi I L Jacobs
- Division of Nuclear Medicine Department of Radiology Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
- School for Mental Health and Neuroscience Alzheimer Centre Limburg Maastricht University Maastricht The Netherlands
| | - Olivia Hampton
- Department of Neurology Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
| | - Edmarie Guzmán-Vélez
- Department of Psychiatry Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
| | - Clara Vila-Castelar
- Department of Psychiatry Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
| | | | - Ana Baena
- Grupo de Neurociencas Universidad de Antioquia Medellín Antioquia Colombia
| | - Arabiye Artola
- Department of Psychiatry Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
| | - Jairo Martinez
- Department of Psychiatry Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
| | - Celina F Pluim
- Department of Psychological and Brain Sciences Boston University Boston Massachusetts USA
- Department of Psychiatry Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
| | - Sergio Alvarez
- Department of Radiology Hospital Pablo Tobon Uribe Medellín Colombia
| | | | | | - Reisa A Sperling
- Department of Neurology Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
- Athinoula A. Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts USA
- Department of Neurology Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| | - Francisco Lopera
- Grupo de Neurociencas Universidad de Antioquia Medellín Antioquia Colombia
| | - Keith A Johnson
- Division of Nuclear Medicine Department of Radiology Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
- Athinoula A. Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts USA
| | - Bradford C Dickerson
- Department of Neurology Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
- Athinoula A. Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts USA
| | - Yakeel T Quiroz
- Department of Psychiatry Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
- Department of Neurology Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
- Grupo de Neurociencas Universidad de Antioquia Medellín Antioquia Colombia
- Athinoula A. Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts USA
| |
Collapse
|
28
|
Sampedro F, Kulisevsky J. Intracortical surface-based MR diffusivity to investigate neurologic and psychiatric disorders: a review. J Neuroimaging 2021; 32:28-35. [PMID: 34506674 DOI: 10.1111/jon.12930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/08/2021] [Accepted: 08/20/2021] [Indexed: 11/29/2022] Open
Abstract
Diffusion tensor imaging (DTI) allows the quantification of water diffusivity within the cerebral cortex. Alterations in cortical mean diffusivity (MD) have been suggested to reflect microstructural damage. Interestingly, microstructural changes can be detected in the absence of macrostructural alterations such as cortical thinning or gray matter volume loss. However, volume-based neuroimaging techniques for the study of cortical MD have shown some limitations in terms of intersubject registration, partial volume correction, and smoothing artifacts. In this review, we summarize how a surface-based approach for the assessment of intracortical MD has not only overcome these technical limitations, but also provided important contributions to the fields of neurology and psychiatry. Since its proposal in 2018, the use of this neuroimaging technique has revealed cortical microstructural alterations in a wide range of clinical contexts, including Alzheimer's disease, Parkinson's disease, schizophrenia, Huntington's disease, multiple sclerosis, amyotrophic lateral sclerosis, and primary progressive aphasia. In most cases, the detection of early intracortical MD alterations preceded the identification of macrostructural changes. Importantly, microstructural damage significantly correlated with cognitive performance and biomarker measures, suggesting a potential role for its use in clinical trials as a sensitive imaging marker of neurodegeneration. Given that DTI is a widely available imaging modality, these encouraging results motivate further research using this novel neuroimaging metric in other clinical contexts. Overall, this technique has shed light into the key role of early cortical degeneration in many diseases where cortical involvement was previously thought to have limited clinical and biological significance.
Collapse
Affiliation(s)
- Frederic Sampedro
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jaime Kulisevsky
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
29
|
Handen B, Clare I, Laymon C, Petersen M, Zaman S, O’Bryant S, Minhas D, Tudorascu D, Brown S, Christian B. Acute Regression in Down Syndrome. Brain Sci 2021; 11:1109. [PMID: 34439728 PMCID: PMC8391552 DOI: 10.3390/brainsci11081109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Acute regression has been reported in some individuals with Down syndrome (DS), typically occurring between the teenage years and mid to late 20s. Characterized by sudden, and often unexplained, reductions in language skills, functional living skills and reduced psychomotor activity, some individuals have been incorrectly diagnosed with Alzheimer's disease (AD). METHODS This paper compares five individuals with DS who previously experienced acute regression with a matched group of 15 unaffected individuals with DS using a set of AD biomarkers. RESULTS While the sample was too small to conduct statistical analyses, findings suggest there are possible meaningful differences between the groups on proteomics biomarkers (e.g., NfL, total tau). Hippocampal, caudate and putamen volumes were slightly larger in the regression group, the opposite of what was hypothesized. A slightly lower amyloid load was found on the PET scans for the regression group, but no differences were noted on tau PET. CONCLUSIONS Some proteomics biomarker findings suggest that individuals with DS who experience acute regression may be at increased risk for AD at an earlier age in comparison to unaffected adults with DS. However, due to the age of the group (mean 38 years), it may be too early to observe meaningful group differences on image-based biomarkers.
Collapse
Affiliation(s)
- Benjamin Handen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15260, USA; (C.L.); (D.M.); (D.T.)
| | - Isabel Clare
- Department of Psychiatry, University of Cambridge, Cambridge CB2 1TN, UK; (I.C.); (S.Z.); (S.B.)
| | - Charles Laymon
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15260, USA; (C.L.); (D.M.); (D.T.)
| | - Melissa Petersen
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (M.P.); (S.O.)
| | - Shahid Zaman
- Department of Psychiatry, University of Cambridge, Cambridge CB2 1TN, UK; (I.C.); (S.Z.); (S.B.)
| | - Sid O’Bryant
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (M.P.); (S.O.)
| | - Davneet Minhas
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15260, USA; (C.L.); (D.M.); (D.T.)
| | - Dana Tudorascu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15260, USA; (C.L.); (D.M.); (D.T.)
| | - Stephanie Brown
- Department of Psychiatry, University of Cambridge, Cambridge CB2 1TN, UK; (I.C.); (S.Z.); (S.B.)
| | - Bradley Christian
- Departments of Medical Physics and Psychiatry, University of Wisconsin, Madison, WI 53706, USA;
| |
Collapse
|
30
|
Solana E, Martinez-Heras E, Montal V, Vilaplana E, Lopez-Soley E, Radua J, Sola-Valls N, Montejo C, Blanco Y, Pulido-Valdeolivas I, Sepúlveda M, Andorra M, Berenguer J, Villoslada P, Martinez-Lapiscina EH, Prados F, Saiz A, Fortea J, Llufriu S. Regional grey matter microstructural changes and volume loss according to disease duration in multiple sclerosis patients. Sci Rep 2021; 11:16805. [PMID: 34413373 PMCID: PMC8376987 DOI: 10.1038/s41598-021-96132-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/03/2021] [Indexed: 01/28/2023] Open
Abstract
The spatio-temporal characteristics of grey matter (GM) impairment in multiple sclerosis (MS) are poorly understood. We used a new surface-based diffusion MRI processing tool to investigate regional modifications of microstructure, and we quantified volume loss in GM in a cohort of patients with MS classified into three groups according to disease duration. Additionally, we investigated the relationship between GM changes with disease severity. We studied 54 healthy controls and 247 MS patients classified regarding disease duration: MS1 (less than 5 years, n = 67); MS2 (5–15 years, n = 107); and MS3 (more than15 years, n = 73). We compared GM mean diffusivity (MD), fractional anisotropy (FA) and volume between groups, and estimated their clinical associations. Regional modifications in diffusion measures (MD and FA) and volume did not overlap early in the disease, and became widespread in later phases. We found higher MD in MS1 group, mainly in the temporal cortex, and volume reduction in deep GM and left precuneus. Additional MD changes were evident in cingulate and occipital cortices in the MS2 group, coupled to volume reductions in deep GM and parietal and frontal poles. Changes in MD and volume extended to more than 80% of regions in MS3 group. Conversely, increments in FA, with very low effect size, were observed in the parietal cortex and thalamus in MS1 and MS2 groups, and extended to the frontal lobe in the later group. MD and GM changes were associated with white matter lesion load and with physical and cognitive disability. Microstructural integrity loss and atrophy present differential spatial predominance early in MS and accrual over time, probably due to distinct pathogenic mechanisms that underlie tissue damage.
Collapse
Affiliation(s)
- Elisabeth Solana
- Center of Neuroimmunology, Laboratory of Advanced Imaging in Neuroimmunological Diseases, Hospital Clinic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Universitat de Barcelona, Barcelona, Spain.
| | - Eloy Martinez-Heras
- Center of Neuroimmunology, Laboratory of Advanced Imaging in Neuroimmunological Diseases, Hospital Clinic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Universitat de Barcelona, Barcelona, Spain.
| | - Victor Montal
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autonoma de Barcelona, Barcelona, Spain.,Centro de Investigacion Biomedica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Eduard Vilaplana
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autonoma de Barcelona, Barcelona, Spain.,Centro de Investigacion Biomedica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Elisabet Lopez-Soley
- Center of Neuroimmunology, Laboratory of Advanced Imaging in Neuroimmunological Diseases, Hospital Clinic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Universitat de Barcelona, Barcelona, Spain
| | - Joaquim Radua
- Imaging of Mood and Anxiety Related Disorders (IMARD) Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Research Networking Center (CIBERSAM), Barcelona, Spain.,Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Centre for Psychiatric Research and Education, Department of Clinical Neuroscience, Karolinska Institutet, Solna, Sweden
| | - Nuria Sola-Valls
- Center of Neuroimmunology, Laboratory of Advanced Imaging in Neuroimmunological Diseases, Hospital Clinic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Universitat de Barcelona, Barcelona, Spain
| | - Carmen Montejo
- Center of Neuroimmunology, Laboratory of Advanced Imaging in Neuroimmunological Diseases, Hospital Clinic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Universitat de Barcelona, Barcelona, Spain
| | - Yolanda Blanco
- Center of Neuroimmunology, Laboratory of Advanced Imaging in Neuroimmunological Diseases, Hospital Clinic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Universitat de Barcelona, Barcelona, Spain
| | - Irene Pulido-Valdeolivas
- Center of Neuroimmunology, Laboratory of Advanced Imaging in Neuroimmunological Diseases, Hospital Clinic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Universitat de Barcelona, Barcelona, Spain
| | - Maria Sepúlveda
- Center of Neuroimmunology, Laboratory of Advanced Imaging in Neuroimmunological Diseases, Hospital Clinic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Universitat de Barcelona, Barcelona, Spain
| | - Magi Andorra
- Center of Neuroimmunology, Laboratory of Advanced Imaging in Neuroimmunological Diseases, Hospital Clinic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Universitat de Barcelona, Barcelona, Spain
| | - Joan Berenguer
- Neuroradiology Section, Radiology Service of the Image Diagnosis Center of the Hospital Clinic de Barcelona, Barcelona, Spain
| | - Pablo Villoslada
- Center of Neuroimmunology, Laboratory of Advanced Imaging in Neuroimmunological Diseases, Hospital Clinic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Universitat de Barcelona, Barcelona, Spain
| | - E H Martinez-Lapiscina
- Center of Neuroimmunology, Laboratory of Advanced Imaging in Neuroimmunological Diseases, Hospital Clinic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Universitat de Barcelona, Barcelona, Spain
| | - Ferran Prados
- E-health Centre, Universitat Oberta de Catalunya, Barcelona, Spain.,Centre for Medical Image Computing (CMIC), Department of Medical Physics and Bioengineering, University College London, London, UK.,NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Institute of Neurology, University College London, London, UK
| | - Albert Saiz
- Center of Neuroimmunology, Laboratory of Advanced Imaging in Neuroimmunological Diseases, Hospital Clinic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Universitat de Barcelona, Barcelona, Spain
| | - Juan Fortea
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autonoma de Barcelona, Barcelona, Spain.,Centro de Investigacion Biomedica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Sara Llufriu
- Center of Neuroimmunology, Laboratory of Advanced Imaging in Neuroimmunological Diseases, Hospital Clinic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
31
|
Rodriguez-Vieitez E, Montal V, Sepulcre J, Lois C, Hanseeuw B, Vilaplana E, Schultz AP, Properzi MJ, Scott MR, Amariglio R, Papp KV, Marshall GA, Fortea J, Johnson KA, Sperling RA, Vannini P. Association of cortical microstructure with amyloid-β and tau: impact on cognitive decline, neurodegeneration, and clinical progression in older adults. Mol Psychiatry 2021; 26:7813-7822. [PMID: 34588623 PMCID: PMC8873001 DOI: 10.1038/s41380-021-01290-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/16/2021] [Accepted: 09/03/2021] [Indexed: 02/07/2023]
Abstract
Noninvasive biomarkers of early neuronal injury may help identify cognitively normal individuals at risk of developing Alzheimer's disease (AD). A recent diffusion-weighted imaging (DWI) method allows assessing cortical microstructure via cortical mean diffusivity (cMD), suggested to be more sensitive than macrostructural neurodegeneration. Here, we aimed to investigate the association of cMD with amyloid-β and tau pathology in older adults, and whether cMD predicts longitudinal cognitive decline, neurodegeneration and clinical progression. The study sample comprised n = 196 cognitively normal older adults (mean[SD] 72.5 [9.4] years; 114 women [58.2%]) from the Harvard Aging Brain Study. At baseline, all participants underwent structural MRI, DWI, 11C-Pittsburgh compound-B-PET, 18F-flortaucipir-PET imaging, and cognitive assessments. Longitudinal measures of Preclinical Alzheimer Cognitive Composite-5 were available for n = 186 individuals over 3.72 (1.96)-year follow-up. Prospective clinical follow-up was available for n = 163 individuals over 3.2 (1.7) years. Surface-based image analysis assessed vertex-wise relationships between cMD, global amyloid-β, and entorhinal and inferior-temporal tau. Multivariable regression, mixed effects models and Cox proportional hazards regression assessed longitudinal cognition, brain structural changes and clinical progression. Tau, but not amyloid-β, was positively associated with cMD in AD-vulnerable regions. Correcting for baseline demographics and cognition, increased cMD predicted steeper cognitive decline, which remained significant after correcting for amyloid-β, thickness, and entorhinal tau; there was a synergistic interaction between cMD and both amyloid-β and tau on cognitive slope. Regional cMD predicted hippocampal atrophy rate, independently from amyloid-β, tau, and thickness. Elevated cMD predicted progression to mild cognitive impairment. Cortical microstructure is a noninvasive biomarker that independently predicts subsequent cognitive decline, neurodegeneration and clinical progression, suggesting utility in clinical trials.
Collapse
Affiliation(s)
- Elena Rodriguez-Vieitez
- grid.38142.3c000000041936754XMassachusetts General Hospital, Harvard Medical School, Boston, MA USA ,grid.509504.d0000 0004 0475 2664Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA USA ,grid.4714.60000 0004 1937 0626Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Victor Montal
- grid.7080.f0000 0001 2296 0625Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain ,grid.418264.d0000 0004 1762 4012Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jorge Sepulcre
- grid.38142.3c000000041936754XMassachusetts General Hospital, Harvard Medical School, Boston, MA USA ,grid.512020.4Gordon Center for Medical Imaging, Boston, MA USA
| | - Cristina Lois
- grid.38142.3c000000041936754XMassachusetts General Hospital, Harvard Medical School, Boston, MA USA ,grid.512020.4Gordon Center for Medical Imaging, Boston, MA USA
| | - Bernard Hanseeuw
- grid.38142.3c000000041936754XMassachusetts General Hospital, Harvard Medical School, Boston, MA USA ,grid.512020.4Gordon Center for Medical Imaging, Boston, MA USA ,grid.7942.80000 0001 2294 713XSaint Luc University Hospital, Université Catholique de Louvain, Brussels, Belgium
| | - Eduard Vilaplana
- grid.7080.f0000 0001 2296 0625Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain ,grid.418264.d0000 0004 1762 4012Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Aaron P. Schultz
- grid.38142.3c000000041936754XMassachusetts General Hospital, Harvard Medical School, Boston, MA USA ,grid.509504.d0000 0004 0475 2664Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA USA
| | - Michael J. Properzi
- grid.38142.3c000000041936754XMassachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Matthew R. Scott
- grid.38142.3c000000041936754XMassachusetts General Hospital, Harvard Medical School, Boston, MA USA ,grid.509504.d0000 0004 0475 2664Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA USA
| | - Rebecca Amariglio
- grid.38142.3c000000041936754XMassachusetts General Hospital, Harvard Medical School, Boston, MA USA ,grid.38142.3c000000041936754XBrigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Kathryn V. Papp
- grid.38142.3c000000041936754XMassachusetts General Hospital, Harvard Medical School, Boston, MA USA ,grid.509504.d0000 0004 0475 2664Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA USA ,grid.38142.3c000000041936754XBrigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Gad A. Marshall
- grid.38142.3c000000041936754XMassachusetts General Hospital, Harvard Medical School, Boston, MA USA ,grid.509504.d0000 0004 0475 2664Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA USA ,grid.38142.3c000000041936754XBrigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Juan Fortea
- grid.7080.f0000 0001 2296 0625Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain ,grid.418264.d0000 0004 1762 4012Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Keith A. Johnson
- grid.38142.3c000000041936754XMassachusetts General Hospital, Harvard Medical School, Boston, MA USA ,grid.512020.4Gordon Center for Medical Imaging, Boston, MA USA
| | - Reisa A. Sperling
- grid.38142.3c000000041936754XMassachusetts General Hospital, Harvard Medical School, Boston, MA USA ,grid.509504.d0000 0004 0475 2664Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA USA ,grid.38142.3c000000041936754XBrigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Patrizia Vannini
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. .,Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA. .,Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|