101
|
Zhuo J, Zhang Y, Liu Y, Liu B, Zhou X, Bartlett PF, Jiang T. New Trajectory of Clinical and Biomarker Changes in Sporadic Alzheimer's Disease. Cereb Cortex 2021; 31:3363-3373. [PMID: 33690839 DOI: 10.1093/cercor/bhab017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/08/2020] [Accepted: 01/18/2021] [Indexed: 12/20/2022] Open
Abstract
Identifying dynamic changes in biomarkers and clinical profiles is essential for understanding the progression of Alzheimer's disease (AD). The relevant studies have primarily relied on patients with autosomal dominant AD; however, relevant studies in sporadic AD are poorly understood. Here, we analyzed longitudinal data from 665 participants (mean follow-up 4.90 ± 2.83 years). By aligning normal cognition (CN) baseline with a clinical diagnosis of mild cognitive impairment (MCI) or AD, we studied the progression of AD using a linear mixed model to estimate the clinical and biomarker changes from stable CN to MCI to AD. The results showed that the trajectory of hippocampal volume and fluorodeoxyglucose (FDG) was consistent with the clinical measures in that they did not follow a hypothetical sigmoid curve but rather showed a slow change in the initial stage and accelerated changes in the later stage from MCI conversion to AD. Dramatic hippocampal atrophy and the ADAS13 increase were, respectively, 2.5 and 1 years earlier than the MCI onset. Besides, cognitively normal people with elevated and normal amyloid showed no significant differences in clinical measures, hippocampal volume, or FDG. These results reveal that pre-MCI to pre-AD may be a better time window for future clinical trial design.
Collapse
Affiliation(s)
- Junjie Zhuo
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,The Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia.,School of Biomedical Engineering, Hainan University, Haikou 570228, China
| | - Yuanchao Zhang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 625014, China
| | - Yong Liu
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,CAS Center for Excellence in Brain Science, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Bing Liu
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,CAS Center for Excellence in Brain Science, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaoqing Zhou
- The Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
| | - Perry F Bartlett
- The Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
| | - Tianzi Jiang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,The Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia.,University of Chinese Academy of Sciences, Beijing 100049, China.,CAS Center for Excellence in Brain Science, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 625014, China
| | | |
Collapse
|
102
|
Shen X, Kuo K, Yang Y, Li H, Chen S, Cui M, Tan L, Dong Q, Yu J. Subtle cognitive impairment as a marker of Alzheimer's pathologies and clinical progression in cognitively normal individuals. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12198. [PMID: 34095433 PMCID: PMC8158163 DOI: 10.1002/dad2.12198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 04/15/2021] [Accepted: 04/21/2021] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Subtle cognitive impairment (SCI) may appear before pathological changes surpass thresholds for abnormality. We aimed to investigate whether SCI could predict Alzheimer's pathologies and advancement. METHODS A total of 816 cognitively normal individuals were enrolled to assess the longitudinal neuropathological and clinical correlates of baseline SCI, via linear mixed-effects and Cox proportional-hazard models. Cross-lagged panel models were used in specific time waves. RESULTS SCI individuals had a faster increase in brain amyloid burden and a higher risk of conversion. They also showed greater rates of cerebrospinal fluid (CSF) phosphorylated tau (p-tau)181 increase and glucose metabolism decrease. In addition, baseline SCI predicted worse clinical progression, whereas multi-domain SCI advanced faster compared to the single domain group. DISCUSSION Baseline SCI could be an imperative prediction indicator of clinical and pathological progression. It enables cognitive measures to be informative at a very early stage and provided objective criteria for high-risk population screening.
Collapse
Affiliation(s)
- Xue‐Ning Shen
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Kevin Kuo
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Yu‐Xiang Yang
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Hong‐Qi Li
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Shi‐Dong Chen
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Mei Cui
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Lan Tan
- Department of NeurologyQingdao Municipal HospitalQingdao UniversityQingdaoChina
| | - Qiang Dong
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Jin‐Tai Yu
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | | |
Collapse
|
103
|
Butt OH, Meeker KL, Wisch JK, Schindler SE, Fagan AM, Benzinger TLS, Cruchaga C, Holtzman DM, Morris JC, Ances BM. Network dysfunction in cognitively normal APOE ε4 carriers is related to subclinical tau. Alzheimers Dement 2021; 18:116-126. [PMID: 34002449 DOI: 10.1002/alz.12375] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/18/2021] [Accepted: 04/16/2021] [Indexed: 11/10/2022]
Abstract
INTRODUCTION Apolipoprotein E (APOE) ε4 allele status is associated with amyloid and tau-related pathological changes related to Alzheimer's disease (AD). However, it is unknown whether brain network changes are related to amyloid beta (Aβ) and/or tau-related pathology in cognitively normal APOE ε4 carriers with subthreshold Aβ accumulation. METHODS Resting state functional connectivity measures of network integrity were evaluated in cognitively normal individuals (n = 121, mean age 76.6 ± 7.8 years, 15% APOE ε4 carriers, 65% female) with minimal Aβ per cerebrospinal fluid (CSF) or amyloid positron emission tomography. RESULTS APOE ε4 carriers had increased lateralized connections relative to callosal connections within the default-mode, memory, and salience networks (P = .02), with significant weighting on linear regression toward CSF total tau (P = .03) and CSF phosphorylated tau at codon 181 (P = .03), but not CSF Aβ42 . DISCUSSION Cognitively normal APOE ε4 carriers with subthreshold amyloid accumulation may have network reorganization associated with tau.
Collapse
Affiliation(s)
- Omar H Butt
- Department of Neurology, Washington University in Saint Louis, Saint Louis, Missouri, USA
| | - Karin L Meeker
- Department of Neurology, Washington University in Saint Louis, Saint Louis, Missouri, USA
| | - Julie K Wisch
- Department of Neurology, Washington University in Saint Louis, Saint Louis, Missouri, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University in Saint Louis, Saint Louis, Missouri, USA
| | - Anne M Fagan
- Department of Neurology, Washington University in Saint Louis, Saint Louis, Missouri, USA.,Knight Alzheimer Disease Research Center, Washington University in Saint Louis, St. Louis, Missouri, USA.,Hope Center for Neurological Disorders, Washington University in Saint Louis, St. Louis, Missouri, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University in Saint Louis, Saint Louis, Missouri, USA
| | - Carlos Cruchaga
- Knight Alzheimer Disease Research Center, Washington University in Saint Louis, St. Louis, Missouri, USA.,Hope Center for Neurological Disorders, Washington University in Saint Louis, St. Louis, Missouri, USA.,Department of Psychiatry, Washington University in Saint Louis, Saint Louis, Missouri, USA
| | - David M Holtzman
- Department of Neurology, Washington University in Saint Louis, Saint Louis, Missouri, USA.,Knight Alzheimer Disease Research Center, Washington University in Saint Louis, St. Louis, Missouri, USA.,Hope Center for Neurological Disorders, Washington University in Saint Louis, St. Louis, Missouri, USA
| | - John C Morris
- Department of Neurology, Washington University in Saint Louis, Saint Louis, Missouri, USA.,Knight Alzheimer Disease Research Center, Washington University in Saint Louis, St. Louis, Missouri, USA.,Hope Center for Neurological Disorders, Washington University in Saint Louis, St. Louis, Missouri, USA
| | - Beau M Ances
- Department of Neurology, Washington University in Saint Louis, Saint Louis, Missouri, USA.,Knight Alzheimer Disease Research Center, Washington University in Saint Louis, St. Louis, Missouri, USA.,Hope Center for Neurological Disorders, Washington University in Saint Louis, St. Louis, Missouri, USA.,Department of Radiology, Washington University in Saint Louis, Saint Louis, Missouri, USA
| |
Collapse
|
104
|
Wisse LEM, de Flores R, Xie L, Das SR, McMillan CT, Trojanowski JQ, Grossman M, Lee EB, Irwin D, Yushkevich PA, Wolk DA. Pathological drivers of neurodegeneration in suspected non-Alzheimer's disease pathophysiology. ALZHEIMERS RESEARCH & THERAPY 2021; 13:100. [PMID: 33990226 PMCID: PMC8122549 DOI: 10.1186/s13195-021-00835-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/26/2021] [Indexed: 11/16/2022]
Abstract
Background Little is known about the heterogeneous etiology of suspected non-Alzheimer’s pathophysiology (SNAP), a group of subjects with neurodegeneration in the absence of β-amyloid. Using antemortem MRI and pathological data, we investigated the etiology of SNAP and the association of neurodegenerative pathologies with structural medial temporal lobe (MTL) measures in β-amyloid-negative subjects. Methods Subjects with antemortem MRI and autopsy data were selected from ADNI (n=63) and the University of Pennsylvania (n=156). Pathological diagnoses and semi-quantitative scores of MTL tau, neuritic plaques, α-synuclein, and TDP-43 pathology and MTL structural MRI measures from antemortem T1-weighted MRI scans were obtained. β-amyloid status (A+/A−) was determined by CERAD score and neurodegeneration status (N+/N−) by hippocampal volume. Results SNAP reflects a heterogeneous group of pathological diagnoses. In ADNI, SNAP (A−N+) had significantly more neuropathological diagnoses than A+N+. In the A− group, tau pathology was associated with hippocampal, entorhinal cortex, and Brodmann area 35 volume/thickness and TDP-43 pathology with hippocampal volume. Conclusion SNAP had a heterogeneous profile with more mixed pathologies than A+N+. Moreover, a role for TDP-43 and tau pathology in driving MTL neurodegeneration in the absence of β-amyloid was supported. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00835-2.
Collapse
Affiliation(s)
- L E M Wisse
- Department of Diagnostic Radiology, Lund University, Remissgatan 4, Room 14-520, 222 42, Lund, Sweden. .,Penn Image Computing and Science Laboratory, Department of Radiology, University of Pennsylvania, Philadelphia, USA. .,Penn Memory Center, Department of Neurology, University of Pennsylvania, Philadelphia, USA.
| | - R de Flores
- Université Normandie, Inserm, Université de Caen-Normandie, Inserm UMR-S U1237, GIP Cyceron, Caen, France
| | - L Xie
- Penn Image Computing and Science Laboratory, Department of Radiology, University of Pennsylvania, Philadelphia, USA.,Penn Memory Center, Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - S R Das
- Penn Memory Center, Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - C T McMillan
- Penn FTD Center, Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - J Q Trojanowski
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, USA
| | - M Grossman
- Penn FTD Center, Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - E B Lee
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, USA
| | - D Irwin
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, USA
| | - P A Yushkevich
- Penn Image Computing and Science Laboratory, Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | - D A Wolk
- Penn Memory Center, Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | | |
Collapse
|
105
|
Knopman DS, Amieva H, Petersen RC, Chételat G, Holtzman DM, Hyman BT, Nixon RA, Jones DT. Alzheimer disease. Nat Rev Dis Primers 2021; 7:33. [PMID: 33986301 PMCID: PMC8574196 DOI: 10.1038/s41572-021-00269-y] [Citation(s) in RCA: 1161] [Impact Index Per Article: 290.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/09/2021] [Indexed: 12/21/2022]
Abstract
Alzheimer disease (AD) is biologically defined by the presence of β-amyloid-containing plaques and tau-containing neurofibrillary tangles. AD is a genetic and sporadic neurodegenerative disease that causes an amnestic cognitive impairment in its prototypical presentation and non-amnestic cognitive impairment in its less common variants. AD is a common cause of cognitive impairment acquired in midlife and late-life but its clinical impact is modified by other neurodegenerative and cerebrovascular conditions. This Primer conceives of AD biology as the brain disorder that results from a complex interplay of loss of synaptic homeostasis and dysfunction in the highly interrelated endosomal/lysosomal clearance pathways in which the precursors, aggregated species and post-translationally modified products of Aβ and tau play important roles. Therapeutic endeavours are still struggling to find targets within this framework that substantially change the clinical course in persons with AD.
Collapse
Affiliation(s)
| | - Helene Amieva
- Inserm U1219 Bordeaux Population Health Center, University of Bordeaux, Bordeaux, France
| | | | - Gäel Chételat
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, Cyceron, Caen, France
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Ralph A Nixon
- Departments of Psychiatry and Cell Biology, New York University Langone Medical Center, New York University, New York, NY, USA
- NYU Neuroscience Institute, New York University Langone Medical Center, New York University, New York, NY, USA
| | - David T Jones
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
106
|
Babulal GM, Roe CM, Stout SH, Rajasekar G, Wisch JK, Benzinger TLS, Morris JC, Ances BM. Depression is Associated with Tau and Not Amyloid Positron Emission Tomography in Cognitively Normal Adults. J Alzheimers Dis 2021; 74:1045-1055. [PMID: 32144985 DOI: 10.3233/jad-191078] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Depression is also common with older age. Alzheimer's disease (AD) studies suggest that both cerebrospinal fluid and positron emission tomography (PET) amyloid biomarkers are associated with more depressive symptoms in cognitively normal older adults. The recent availability of tau radiotracers offers the ability to examine in vivo tauopathy. It is unclear if the tau biomarker is associated with depression diagnosis. OBJECTIVE We examined if tau and amyloid imaging were associated with a depression diagnosis among cognitively normal adults (Clinical Dementia Rating = 0) and whether antidepressants modified this relationship. METHODS Among 301 participants, logistic regression models evaluated whether in vivo PET tau was associated with depression, while another model tested the interaction between PET tau and antidepressant use. A second set of models substituted PET amyloid for PET tau. A diagnosis of depression (yes/no) was made during an annual clinical assessment by a clinician. Antidepressant use (yes/no) was determined by comparing medications the participants used to a list of 30 commonly used antidepressants. All models adjusted for age, sex, education, race, and apolipoprotein ɛ4. Similar models explored the association between the biomarkers and depressive symptoms. RESULTS Participants with elevated tau were twice as likely to be depressed. Antidepressant use modified this relationship where participants with elevated tau who were taking antidepressants had greater odds of being depressed. Relatedly, elevated amyloid was not associated with depression. CONCLUSIONS Our results demonstrate that tau, not amyloid, was associated with a depression diagnosis. Additionally, antidepressant use interacts with tau to increase the odds of depression among cognitively normal adults.
Collapse
Affiliation(s)
- Ganesh M Babulal
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Catherine M Roe
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sarah H Stout
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ganesh Rajasekar
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Julie K Wisch
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.,Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.,Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Physical Therapy, Washington University School of Medicine, St. Louis, MO, USA.,Department of Occupational Therapy, Washington University School of Medicine, St. Louis, MO, USA
| | - Beau M Ances
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
107
|
Sutoko S, Masuda A, Kandori A, Sasaguri H, Saito T, Saido TC, Funane T. Early Identification of Alzheimer's Disease in Mouse Models: Application of Deep Neural Network Algorithm to Cognitive Behavioral Parameters. iScience 2021; 24:102198. [PMID: 33733064 PMCID: PMC7937558 DOI: 10.1016/j.isci.2021.102198] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 01/12/2021] [Accepted: 02/11/2021] [Indexed: 01/15/2023] Open
Abstract
Alzheimer's disease (AD) is a worldwide burden. Diagnosis is complicated by the fact that AD is asymptomatic at an early stage. Studies using AD-modeled animals offer important and useful insights. Here, we classified mice with a high risk of AD at a preclinical stage by using only their behaviors. Wild-type and knock-in AD-modeled (App NL-G-F/NL-G-F ) mice were raised, and their cognitive behaviors were assessed in an automated monitoring system. The classification utilized a machine learning method, i.e., a deep neural network, together with optimized stepwise feature selection and cross-validation. The AD risk could be identified on the basis of compulsive and learning behaviors (89.3% ± 9.8% accuracy) shown by AD-modeled mice in the early age (i.e., 8-12 months old) when the AD symptomatic cognitions were relatively underdeveloped. This finding reveals the advantage of machine learning in unveiling the importance of compulsive and learning behaviors for early AD diagnosis in mice.
Collapse
Affiliation(s)
- Stephanie Sutoko
- Hitachi, Ltd, Research and Development Group, Center for Exploratory Research, Kokubunji, Tokyo 185-8601, Japan
| | - Akira Masuda
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan
- Organization for Research Initiatives and Development, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| | - Akihiko Kandori
- Hitachi, Ltd, Research and Development Group, Center for Exploratory Research, Kokubunji, Tokyo 185-8601, Japan
| | - Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan
| | - Tsukasa Funane
- Hitachi, Ltd, Research and Development Group, Center for Exploratory Research, Kokubunji, Tokyo 185-8601, Japan
| |
Collapse
|
108
|
Keshavan A, Pannee J, Karikari TK, Rodriguez JL, Ashton NJ, Nicholas JM, Cash DM, Coath W, Lane CA, Parker TD, Lu K, Buchanan SM, Keuss SE, James SN, Murray-Smith H, Wong A, Barnes A, Dickson JC, Heslegrave A, Portelius E, Richards M, Fox NC, Zetterberg H, Blennow K, Schott JM. Population-based blood screening for preclinical Alzheimer's disease in a British birth cohort at age 70. Brain 2021; 144:434-449. [PMID: 33479777 PMCID: PMC7940173 DOI: 10.1093/brain/awaa403] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/10/2020] [Accepted: 09/17/2020] [Indexed: 11/14/2022] Open
Abstract
Alzheimer's disease has a preclinical stage when cerebral amyloid-β deposition occurs before symptoms emerge, and when amyloid-β-targeted therapies may have maximum benefits. Existing amyloid-β status measurement techniques, including amyloid PET and CSF testing, are difficult to deploy at scale, so blood biomarkers are increasingly considered for screening. We compared three different blood-based techniques-liquid chromatography-mass spectrometry measures of plasma amyloid-β, and single molecule array (Simoa) measures of plasma amyloid-β and phospho-tau181-to detect cortical 18F-florbetapir amyloid PET positivity (defined as a standardized uptake value ratio of >0.61 between a predefined cortical region of interest and eroded subcortical white matter) in dementia-free members of Insight 46, a substudy of the population-based British 1946 birth cohort. We used logistic regression models with blood biomarkers as predictors of amyloid PET status, with or without age, sex and APOE ε4 carrier status as covariates. We generated receiver operating characteristics curves and quantified areas under the curves to compare the concordance of the different blood tests with amyloid PET. We determined blood test cut-off points using Youden's index, then estimated numbers needed to screen to obtain 100 amyloid PET-positive individuals. Of the 502 individuals assessed, 441 dementia-free individuals with complete data were included; 82 (18.6%) were amyloid PET-positive. The area under the curve for amyloid PET status using a base model comprising age, sex and APOE ε4 carrier status was 0.695 (95% confidence interval: 0.628-0.762). The two best-performing Simoa plasma biomarkers were amyloid-β42/40 (0.620; 0.548-0.691) and phospho-tau181 (0.707; 0.646-0.768), but neither outperformed the base model. Mass spectrometry plasma measures performed significantly better than any other measure (amyloid-β1-42/1-40: 0.817; 0.770-0.864 and amyloid-β composite: 0.820; 0.775-0.866). At a cut-off point of 0.095, mass spectrometry measures of amyloid-β1-42/1-40 detected amyloid PET positivity with 86.6% sensitivity and 71.9% specificity. Without screening, to obtain 100 PET-positive individuals from a population with similar amyloid PET positivity prevalence to Insight 46, 543 PET scans would need to be performed. Screening using age, sex and APOE ε4 status would require 940 individuals, of whom 266 would proceed to scan. Using mass spectrometry amyloid-β1-42/1-40 alone would reduce these numbers to 623 individuals and 243 individuals, respectively. Across a theoretical range of amyloid PET positivity prevalence of 10-50%, mass spectrometry measures of amyloid-β1-42/1-40 would consistently reduce the numbers proceeding to scans, with greater cost savings demonstrated at lower prevalence.
Collapse
Affiliation(s)
- Ashvini Keshavan
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Josef Pannee
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Thomas K Karikari
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Juan Lantero Rodriguez
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Nicholas J Ashton
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- National Institute for Health Research Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation Trust, London, UK
| | - Jennifer M Nicholas
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, University of London, London, UK
| | - David M Cash
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - William Coath
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Christopher A Lane
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Thomas D Parker
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Kirsty Lu
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Sarah M Buchanan
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Sarah E Keuss
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | - Heidi Murray-Smith
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Andrew Wong
- MRC Unit for Lifelong Health and Ageing at UCL, London, UK
| | - Anna Barnes
- Institute of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, London, UK
| | - John C Dickson
- Institute of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, London, UK
| | - Amanda Heslegrave
- UK Dementia Research Institute Fluid Biomarkers Laboratory, UK DRI at UCL, London, UK
| | - Erik Portelius
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | | | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute Fluid Biomarkers Laboratory, UK DRI at UCL, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Jonathan M Schott
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
109
|
Fischer FU, Wolf D, Tüscher O, Fellgiebel A. Structural Network Efficiency Predicts Resilience to Cognitive Decline in Elderly at Risk for Alzheimer's Disease. Front Aging Neurosci 2021; 13:637002. [PMID: 33692682 PMCID: PMC7937862 DOI: 10.3389/fnagi.2021.637002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/25/2021] [Indexed: 01/10/2023] Open
Abstract
Introduction: Functional imaging studies have demonstrated the recruitment of additional neural resources as a possible mechanism to compensate for age and Alzheimer's disease (AD)-related cerebral pathology, the efficacy of which is potentially modulated by underlying structural network connectivity. Additionally, structural network efficiency (SNE) is associated with intelligence across the lifespan, which is a known factor for resilience to cognitive decline. We hypothesized that SNE may be a surrogate of the physiological basis of resilience to cognitive decline in elderly persons without dementia and with age- and AD-related cerebral pathology.Methods: We included 85 cognitively normal elderly subjects or mild cognitive impairment (MCI) patients submitted to baseline diffusion imaging, liquor specimens, amyloid-PET and longitudinal cognitive assessments. SNE was calculated from baseline MRI scans using fiber tractography and graph theory. Mixed linear effects models were estimated to investigate the association of higher resilience to cognitive decline with higher SNE and the modulation of this association by increased cerebral amyloid, liquor tau or WMHV. Results: For the majority of cognitive outcome measures, higher SNE was associated with higher resilience to cognitive decline (p-values: 0.011-0.039). Additionally, subjects with higher SNE showed more resilience to cognitive decline at higher cerebral amyloid burden (p-values: <0.001-0.036) and lower tau levels (p-values: 0.002-0.015).Conclusion: These results suggest that SNE to some extent may quantify the physiological basis of resilience to cognitive decline most effective at the earliest stages of AD, namely at increased amyloid burden and before increased tauopathy.
Collapse
Affiliation(s)
- Florian U. Fischer
- Department of Psychiatry and Psychotherapy, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
- Center for Mental Health in Old Age, Landeskrankenhaus (AöR), Mainz, Germany
| | - Dominik Wolf
- Department of Psychiatry and Psychotherapy, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
- Center for Mental Health in Old Age, Landeskrankenhaus (AöR), Mainz, Germany
| | - Oliver Tüscher
- Department of Psychiatry and Psychotherapy, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
- Leibniz Institute for Resilience Research (LIR), Mainz, Germany
| | - Andreas Fellgiebel
- Department of Psychiatry and Psychotherapy, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
- Center for Mental Health in Old Age, Landeskrankenhaus (AöR), Mainz, Germany
| | | |
Collapse
|
110
|
Belloy ME, Napolioni V, Han SS, Le Guen Y, Greicius MD. Association of Klotho-VS Heterozygosity With Risk of Alzheimer Disease in Individuals Who Carry APOE4. JAMA Neurol 2021; 77:849-862. [PMID: 32282020 DOI: 10.1001/jamaneurol.2020.0414] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Importance Identification of genetic factors that interact with the apolipoprotein e4 (APOE4) allele to reduce risk for Alzheimer disease (AD) would accelerate the search for new AD drug targets. Klotho-VS heterozygosity (KL-VSHET+ status) protects against aging-associated phenotypes and cognitive decline, but whether it protects individuals who carry APOE4 from AD remains unclear. Objectives To determine if KL-VSHET+ status is associated with reduced AD risk and β-amyloid (Aβ) pathology in individuals who carry APOE4. Design, Setting, and Participants This study combined 25 independent case-control, family-based, and longitudinal AD cohorts that recruited referred and volunteer participants and made data available through public repositories. Analyses were stratified by APOE4 status. Three cohorts were used to evaluate conversion risk, 1 provided longitudinal measures of Aβ CSF and PET, and 3 provided cross-sectional measures of Aβ CSF. Genetic data were available from high-density single-nucleotide variant microarrays. All data were collected between September 2015 and September 2019 and analyzed between April 2019 and December 2019. Main Outcomes and Measures The risk of AD was evaluated through logistic regression analyses under a case-control design. The risk of conversion to mild cognitive impairment (MCI) or AD was evaluated through competing risks regression. Associations with Aβ, measured from cerebrospinal fluid (CSF) or brain positron emission tomography (PET), were evaluated using linear regression and mixed-effects modeling. Results Of 36 530 eligible participants, 13 782 were excluded for analysis exclusion criteria or refusal to participate. Participants were men and women aged 60 years and older who were non-Hispanic and of Northwestern European ancestry and had been diagnosed as being cognitively normal or having MCI or AD. The sample included 20 928 participants in case-control studies, 3008 in conversion studies, 556 in Aβ CSF regression analyses, and 251 in PET regression analyses. The genotype KL-VSHET+ was associated with reduced risk for AD in individuals carrying APOE4 who were 60 years or older (odds ratio, 0.75 [95% CI, 0.67-0.84]; P = 7.4 × 10-7), and this was more prominent at ages 60 to 80 years (odds ratio, 0.69 [95% CI, 0.61-0.79]; P = 3.6 × 10-8). Additionally, control participants carrying APOE4 with KL-VS heterozygosity were at reduced risk of converting to MCI or AD (hazard ratio, 0.64 [95% CI, 0.44-0.94]; P = .02). Finally, in control participants who carried APOE4 and were aged 60 to 80 years, KL-VS heterozygosity was associated with higher Aβ in CSF (β, 0.06 [95% CI, 0.01-0.10]; P = .03) and lower Aβ on PET scans (β, -0.04 [95% CI, -0.07 to -0.00]; P = .04). Conclusions and Relevance The genotype KL-VSHET+ is associated with reduced AD risk and Aβ burden in individuals who are aged 60 to 80 years, cognitively normal, and carrying APOE4. Molecular pathways associated with KL merit exploration for novel AD drug targets. The KL-VS genotype should be considered in conjunction with the APOE genotype to refine AD prediction models used in clinical trial enrichment and personalized genetic counseling.
Collapse
Affiliation(s)
- Michael E Belloy
- Department of Neurology and Neurological Sciences, Functional Imaging in Neuropsychiatric Disorders (FIND) Lab, Stanford University, Stanford, California
| | - Valerio Napolioni
- Department of Neurology and Neurological Sciences, Functional Imaging in Neuropsychiatric Disorders (FIND) Lab, Stanford University, Stanford, California
| | - Summer S Han
- Department of Neurosurgery, Stanford University, Stanford, California.,Quantitative Sciences Unit, Stanford Medicine, Stanford, California
| | - Yann Le Guen
- Department of Neurology and Neurological Sciences, Functional Imaging in Neuropsychiatric Disorders (FIND) Lab, Stanford University, Stanford, California
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Functional Imaging in Neuropsychiatric Disorders (FIND) Lab, Stanford University, Stanford, California
| | | |
Collapse
|
111
|
Sperling RA, Donohue MC, Raman R, Sun CK, Yaari R, Holdridge K, Siemers E, Johnson KA, Aisen PS. Association of Factors With Elevated Amyloid Burden in Clinically Normal Older Individuals. JAMA Neurol 2021; 77:735-745. [PMID: 32250387 DOI: 10.1001/jamaneurol.2020.0387] [Citation(s) in RCA: 211] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Importance The Anti-Amyloid Treatment in Asymptomatic Alzheimer disease (A4) Study is an ongoing prevention trial in clinically normal older individuals with evidence of elevated brain amyloid. The large number of participants screened with amyloid positron emission tomography (PET) and standardized assessments provides an unprecedented opportunity to evaluate factors associated with elevated brain amyloid. Objective To investigate the association of elevated amyloid with demographic and lifestyle factors, apolipoprotein E (APOE), neuropsychological testing, and self- and study partner reports of cognitive function. Design, Setting, and Participants This cross-sectional study included screening data in the Anti-Amyloid Treatment in Asymptomatic Alzheimer Disease (A4) Study collected from April 2014 to December 2017 and classified by amyloid status. Data were was analyzed from 2018 to 2019 across 67 sites in the US, Canada, Australia, and Japan and included 4486 older individuals (age 65-85 years) who were eligible for amyloid PET (clinically normal [Clinical Dementia Rating = 0] and cognitively unimpaired [Mini-Mental State Examination score, ≥25; logical memory IIa 6-18]). Main Outcomes and Measures Screening demographics, lifestyle variables, APOE genotyping, and cognitive testing (Preclinical Alzheimer Cognitive Composite), self- and study partner reports of high-level daily cognitive function (Cognitive Function Index). Florbetapir amyloid PET imaging was used to classify participants as having elevated amyloid (Aβ+) or not having elevated amyloid (Aβ-). Results Amyloid PET results were acquired for 4486 participants (mean [SD] age, 71.29 [4.67] years; 2647 women [59%]), with 1323 (29.5%) classified as Aβ+. Aβ+ participants were slightly older than Aβ-, with no observed differences in sex, education, marital or retirement status, or any self-reported lifestyle factors. Aβ+ participants were more likely to have a family history of dementia (3320 Aβ+ [74%] vs 3050 Aβ- [68%]) and at least 1 APOE ε4 allele (2602 Aβ+ [58%] vs 1122 Aβ- [25%]). Aβ+ participants demonstrated worse performance on screening Preclinical Alzheimer Cognitive Composite results and reported higher change scores on the Cognitive Function Index. Conclusions and Relevance Among a large group of older individuals screening for an Alzheimer disease (AD) prevention trial, elevated brain amyloid was associated with family history and APOE ε4 allele but not with multiple other previously reported risk factors for AD. Elevated amyloid was associated with lower test performance results and increased reports of subtle recent declines in daily cognitive function. These results support the hypothesis that elevated amyloid represents an early stage in the Alzheimer continuum and demonstrate the feasibility of enrolling these high-risk participants in secondary prevention trials aimed at slowing cognitive decline during the preclinical stages of AD.
Collapse
Affiliation(s)
- Reisa A Sperling
- Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Harvard Aging Brain Study, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Michael C Donohue
- Alzheimer Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego
| | - Rema Raman
- Alzheimer Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego
| | - Chung-Kai Sun
- Alzheimer Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego
| | - Roy Yaari
- Eli Lilly & Co, Indianapolis, Indiana
| | | | - Eric Siemers
- Eli Lilly & Co, Indianapolis, Indiana.,Siemers Integration LLC
| | - Keith A Johnson
- Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Harvard Aging Brain Study, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Paul S Aisen
- Alzheimer Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego
| | | |
Collapse
|
112
|
Arbaciauskaite M, Lei Y, Cho YK. High-specificity antibodies and detection methods for quantifying phosphorylated tau from clinical samples. Antib Ther 2021; 4:34-44. [PMID: 33928234 PMCID: PMC7944500 DOI: 10.1093/abt/tbab004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/14/2021] [Accepted: 02/05/2021] [Indexed: 12/14/2022] Open
Abstract
The ability to measure total and phosphorylated tau levels in clinical samples is transforming the detection of Alzheimer’s disease (AD) and other neurodegenerative diseases. In particular, recent reports indicate that accurate detection of low levels of phosphorylated tau (p-tau) in plasma provides a reliable biomarker of AD long before sensing memory loss. Therefore, the diagnosis and monitoring of neurodegenerative diseases progression using blood samples is becoming a reality. These major advances were achieved by using antibodies specific to p-tau as well as sophisticated high-sensitivity immunoassay platforms. This review focuses on these enabling advances in high-specificity antibody development, engineering, and novel signal detection methods. We will draw insights from structural studies on p-tau antibodies, engineering efforts to improve their binding properties, and efforts to validate their specificity. A comprehensive survey of high-sensitivity p-tau immunoassay platforms along with sensitivity limits will be provided. We conclude that although robust approaches for detecting certain p-tau species have been established, systematic efforts to validate antibodies for assay development is still needed for the recognition of biomarkers for AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Monika Arbaciauskaite
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Yu Lei
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Yong Ku Cho
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
113
|
Pelkmans W, Legdeur N, Ten Kate M, Barkhof F, Yaqub MM, Holstege H, van Berckel BNM, Scheltens P, van der Flier WM, Visser PJ, Tijms BM. Amyloid-β, cortical thickness, and subsequent cognitive decline in cognitively normal oldest-old. Ann Clin Transl Neurol 2021; 8:348-358. [PMID: 33421355 PMCID: PMC7886045 DOI: 10.1002/acn3.51273] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 12/14/2022] Open
Abstract
Objective To investigate the relationship between amyloid‐β (Aβ) deposition and markers of brain structure on cognitive decline in oldest‐old individuals with initial normal cognition. Methods We studied cognitive functioning in four domains at baseline and change over time in fifty‐seven cognitively intact individuals from the EMIF‐AD 90+ study. Predictors were Aβ status determined by [18F]‐flutemetamol PET (normal = Aβ − vs. abnormal = Aβ+), cortical thickness in 34 regions and hippocampal volume. Mediation analyses were performed to test whether effects of Aβ on cognitive decline were mediated by atrophy of specific anatomical brain areas. Results Subjects had a mean age of 92.7 ± 2.9 years, of whom 19 (33%) were Aβ+. Compared to Aβ−, Aβ+ individuals showed steeper decline on memory (β ± SE = −0.26 ± 0.09), and processing speed (β ± SE = −0.18 ± 0.08) performance over 1.5 years (P < 0.05). Furthermore, medial and lateral temporal lobe atrophy was associated with steeper decline in memory and language across individuals. Mediation analyses revealed that part of the memory decline observed in Aβ+ individuals was mediated through parahippocampal atrophy. Interpretation These results show that Aβ abnormality even in the oldest old with initially normal cognition is not part of normal aging, but is associated with a decline in cognitive functioning. Other pathologies may also contribute to decline in the oldest old as cortical thickness predicted cognitive decline similarly in individuals with and without Aβ pathology.
Collapse
Affiliation(s)
- Wiesje Pelkmans
- Alzheimer Center Amsterdam, Department of Neurology I Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Nienke Legdeur
- Alzheimer Center Amsterdam, Department of Neurology I Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Mara Ten Kate
- Alzheimer Center Amsterdam, Department of Neurology I Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Queen Square Institute of Neurology and Centre for Medical Image Computing, UCL, London, UK
| | - Maqsood M Yaqub
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Henne Holstege
- Alzheimer Center Amsterdam, Department of Neurology I Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Department of Clinical Genetics, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Bart N M van Berckel
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology I Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology I Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Department of Epidemiology & Biostatistics, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Department of Neurology I Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Department of Neurology I Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
114
|
Dunne RA, Aarsland D, O’Brien JT, Ballard C, Banerjee S, Fox NC, Isaacs JD, Underwood BR, Perry RJ, Chan D, Dening T, Thomas AJ, Schryer J, Jones AM, Evans AR, Alessi C, Coulthard EJ, Pickett J, Elton P, Jones RW, Mitchell S, Hooper N, Kalafatis C, Rasmussen JGC, Martin H, Schott JM, Burns A. Mild cognitive impairment: the Manchester consensus. Age Ageing 2021; 50:72-80. [PMID: 33197937 PMCID: PMC7793599 DOI: 10.1093/ageing/afaa228] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Indexed: 11/14/2022] Open
Abstract
Given considerable variation in diagnostic and therapeutic practice, there is a need for national guidance on the use of neuroimaging, fluid biomarkers, cognitive testing, follow-up and diagnostic terminology in mild cognitive impairment (MCI). MCI is a heterogenous clinical syndrome reflecting a change in cognitive function and deficits on neuropsychological testing but relatively intact activities of daily living. MCI is a risk state for further cognitive and functional decline with 5-15% of people developing dementia per year. However, ~50% remain stable at 5 years and in a minority, symptoms resolve over time. There is considerable debate about whether MCI is a useful clinical diagnosis, or whether the use of the term prevents proper inquiry (by history, examination and investigations) into underlying causes of cognitive symptoms, which can include prodromal neurodegenerative disease, other physical or psychiatric illness, or combinations thereof. Cognitive testing, neuroimaging and fluid biomarkers can improve the sensitivity and specificity of aetiological diagnosis, with growing evidence that these may also help guide prognosis. Diagnostic criteria allow for a diagnosis of Alzheimer's disease to be made where MCI is accompanied by appropriate biomarker changes, but in practice, such biomarkers are not available in routine clinical practice in the UK. This would change if disease-modifying therapies became available and required a definitive diagnosis but would present major challenges to the National Health Service and similar health systems. Significantly increased investment would be required in training, infrastructure and provision of fluid biomarkers and neuroimaging. Statistical techniques combining markers may provide greater sensitivity and specificity than any single disease marker but their practical usefulness will depend on large-scale studies to ensure ecological validity and that multiple measures, e.g. both cognitive tests and biomarkers, are widely available for clinical use. To perform such large studies, we must increase research participation amongst those with MCI.
Collapse
Affiliation(s)
| | - Dag Aarsland
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - John T O’Brien
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge CB2 0SP, UK
| | | | | | - Nick C Fox
- University College London, London WC1E 6BT, UK
| | - Jeremy D Isaacs
- St George's University Hospitals NHS Foundation Trust, London SW17 0QT, UK
| | - Benjamin R Underwood
- Gnodde Goldman Sachs Translational Neuroscience Unit, Cambridgeshire and Peterborough Foundation Trust, Cambridge, UK
| | | | - Dennis Chan
- Institute of Cognitive Neuroscience, UCL, London, UK
| | - Tom Dening
- Division of Psychiatry and Applied Psychology, University of Nottingham, Nottingham, UK
| | - Alan J Thomas
- Newcastle University, Gateshead Health NHS Foundation Trust, Newcastle, UK
| | | | | | | | | | | | | | - Peter Elton
- Greater Manchester and Eastern Cheshire Strategic Clinical Network, Manchester M1 3BN UK
| | - Roy W Jones
- The Research Institute for the Care of Older People, Royal United Hospital, Combe Park, Bath BA1 3NG, UK
| | | | - Nigel Hooper
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Chris Kalafatis
- Clinical Trials/S&L Care Home Intervention Team, South London and Maudsley NHS Foundation Trust, Department of Old Age Psychiatry, IOPPN, London SE5 8AF, UK
| | | | - Helen Martin
- Greater Manchester Dementia Research Centre, Palliative Care Lead, Dementia United, Greater Manchester Health and Social Care Partnership, Manchester M! 2BN, UK
| | - Jonathan M Schott
- Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Alistair Burns
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
| |
Collapse
|
115
|
Rådestig MA, Skoog J, Zetterberg H, Kern J, Zettergren A, Sacuiu S, Waern M, Wetterberg H, Blennow K, Skoog I, Kern S. Cognitive Performance and Cerebrospinal Fluid Markers in Preclinical Alzheimer's Disease: Results from the Gothenburg H70 Birth Cohort Studies. J Alzheimers Dis 2021; 79:225-235. [PMID: 33216028 DOI: 10.3233/jad-200751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND We have previously shown that older adults with preclinical Alzheimer's disease (AD) pathology in cerebrospinal fluid (CSF) had slightly worse performance in Mini-Mental State Examination (MMSE) than participants without preclinical AD pathology. OBJECTIVE We therefore aimed to compare performance on neurocognitive tests in a population-based sample of 70-year-olds with and without CSF AD pathology. METHODS The sample was derived from the population-based Gothenburg H70 Birth Cohort Studies in Sweden. Participants (n = 316, 70 years old) underwent comprehensive cognitive examinations, and CSF Aβ-42, Aβ-40, T-tau, and P-tau concentrations were measured. Participants were classified according to the ATN system, and according to their Clinical Dementia Rating (CDR) score. Cognitive performance was examined in the CSF amyloid, tau, and neurodegeneration (ATN) categories. RESULTS Among participants with CDR 0 (n = 259), those with amyloid (A+) and/or tau pathology (T+, N+) showed similar performance on most cognitive tests compared to participants with A-T-N-. Participants with A-T-N+ performed worse in memory (Supra span (p = 0.003), object Delayed (p = 0.042) and Immediate recall (p = 0.033)). Among participants with CDR 0.5 (n = 57), those with amyloid pathology (A+) scored worse in category fluency (p = 0.003). CONCLUSION Cognitively normal participants with amyloid and/or tau pathology performed similarly to those without any biomarker evidence of preclinical AD in most cognitive domains, with the exception of slightly poorer memory performance in A-T-N+. Our study suggests that preclinical AD biomarkers are altered before cognitive decline.
Collapse
Affiliation(s)
- Maya Arvidsson Rådestig
- Center for Ageing and Health (AgeCap) at the University of Gothenburg, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Johan Skoog
- Center for Ageing and Health (AgeCap) at the University of Gothenburg, Mölndal, Sweden.,Department of Psychology, University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,UCL Institute of Neurology (H.Z.), Queen Square, London, United Kingdom.,The UK Dementia Research Institute at UCL, London, United Kingdom
| | - Jürgen Kern
- Center for Ageing and Health (AgeCap) at the University of Gothenburg, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Anna Zettergren
- Center for Ageing and Health (AgeCap) at the University of Gothenburg, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Simona Sacuiu
- Center for Ageing and Health (AgeCap) at the University of Gothenburg, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Margda Waern
- Center for Ageing and Health (AgeCap) at the University of Gothenburg, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Hanna Wetterberg
- Center for Ageing and Health (AgeCap) at the University of Gothenburg, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Ingmar Skoog
- Center for Ageing and Health (AgeCap) at the University of Gothenburg, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Silke Kern
- Center for Ageing and Health (AgeCap) at the University of Gothenburg, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| |
Collapse
|
116
|
Kvello-Alme M, Bråthen G, White LR, Sando SB. Time to Diagnosis in Young Onset Alzheimer's Disease: A Population-Based Study from Central Norway. J Alzheimers Dis 2021; 82:965-974. [PMID: 34120901 PMCID: PMC8461696 DOI: 10.3233/jad-210090] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2021] [Indexed: 11/15/2022]
Abstract
BACKGROUND Young onset dementia is associated with a longer time to diagnosis compared to late onset dementia. Earlier publications have indicated that atypical presentation is a key contributing factor to the diagnostic delay. Our hypothesis was that even the most common presentation of Alzheimer's disease is associated with a substantial diagnostic delay in patients < 65 years. OBJECTIVE To determine the time to diagnosis, and time lags in the diagnostic pathway in typical young onset Alzheimer's disease in central Norway. METHODS The main sources of patients were the databases at the Department of Neurology, University Hospital of Trondheim (St. Olav's Hospital), and Department of Psychiatry, Levanger Hospital. Other sources included key persons in the communities, collaborating hospital departments examining patients with suspected cognitive impairment, and review of hospital records of all three hospitals in the area. Information on the time lags, and the clinical assessment, including the use of biomarkers, was collected from hospital notes. Caregivers were interviewed by telephone. RESULTS Time from first symptom to diagnosis in typical young onset Alzheimer's disease was 5.5 years (n = 223, SD 2.8). Time from onset to contact with healthcare services (usually a general practitioner) was 3.4 years (SD 2.3). Time from contact with healthcare services to the first visit at a hospital was 10.3 months (SD 15.5). Time from first visit at a hospital to diagnosis was 14.8 months (SD 22.6). The analysis of cerebrospinal fluid core biomarkers was performed after 8.3 months (SD 20.9). CONCLUSION Typical Alzheimer's disease is associated with a substantial diagnostic delay in younger patients. Raising public awareness, and education of healthcare professionals on the aspects of young onset Alzheimer's disease is warranted. CSF core biomarkers should be performed earlier in the hospital evaluation process.
Collapse
Affiliation(s)
- Marte Kvello-Alme
- Department of Neuromedicine and Movement Science (INB), Faculty of Medicine and Health Sciences, NTNU, Trondheim, Norway
- Department of Psychiatry, Nord-Trøndelag Hospital Trust, Levanger Hospital, Levanger, Norway
| | - Geir Bråthen
- Department of Neuromedicine and Movement Science (INB), Faculty of Medicine and Health Sciences, NTNU, Trondheim, Norway
- Department of Neurology and Clinical Neurophysiology, University Hospital of Trondheim, Trondheim, Norway
| | - Linda R. White
- Department of Neuromedicine and Movement Science (INB), Faculty of Medicine and Health Sciences, NTNU, Trondheim, Norway
| | - Sigrid Botne Sando
- Department of Neuromedicine and Movement Science (INB), Faculty of Medicine and Health Sciences, NTNU, Trondheim, Norway
- Department of Neurology and Clinical Neurophysiology, University Hospital of Trondheim, Trondheim, Norway
| |
Collapse
|
117
|
Dehghani C, Frost S, Jayasena R, Fowler C, Masters CL, Kanagasingam Y, Jiao H, Lim JKH, Chinnery HR, Downie LE. Morphometric Changes to Corneal Dendritic Cells in Individuals With Mild Cognitive Impairment. Front Neurosci 2020; 14:556137. [PMID: 33362451 PMCID: PMC7755610 DOI: 10.3389/fnins.2020.556137] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 11/09/2020] [Indexed: 11/13/2022] Open
Abstract
Purpose There has been increasing interest in identifying non-invasive, imaging biomarkers for neurodegenerative disorders of the central nervous system (CNS). The aim of this proof-of-concept study was to investigate whether corneal sensory nerve and dendritic cell (DC) parameters, captured using in vivo confocal microscopy (IVCM), are altered in individuals with mild cognitive impairment (MCI) and Alzheimer’s disease (AD). Methods Fifteen participants were recruited from the Australian Imaging Biomarkers and Lifestyle (AIBL) study in Melbourne, VIC, Australia. The cohort consisted of cognitively normal (CN) individuals (n = 5), and those with MCI (n = 5) and AD (n = 5). Participants underwent a slit lamp examination of the anterior segment, followed by corneal imaging using laser-scanning in vivo confocal microscopy (IVCM) of the central and inferior whorl regions. Corneal DC density, field area, perimeter, circularity index, aspect ratio, and roundness were quantified using Image J. Quantitative data were derived for corneal nerve parameters, including nerve fiber length (CNFL), fiber density (CNFD), branch density (CNBD), and diameter. Results Corneal DC field area and perimeter were greater in individuals with MCI, relative to CN controls, in both the central and inferior whorl regions (p < 0.05 for all comparisons). In addition, corneal DCs in the whorl region of MCI eyes had lower circularity and roundness indices and a higher aspect ratio relative to CNs (p < 0.05 for all comparisons). DC density was similar across participant groups in both corneal regions. There was a trend toward lower quantitative parameters for corneal nerve architecture in the AD and MCI groups compared with CN participants, however, the inter-group differences did not reach statistical significance. Central corneal nerve diameters were similar between groups. Conclusion This study is the first to report morphological differences in corneal DCs in humans with MCI. These differences were evident in both the central and mid-peripheral cornea, and in the absence of significant nerve abnormalities or a difference in DC density. These findings justify future large-scale studies to assess the utility of corneal IVCM and DC analysis for identifying early stage pathology in neurodegenerative disorders of the CNS.
Collapse
Affiliation(s)
- Cirous Dehghani
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia.,CSIRO, Australian e-Health Research Centre (AEHRC), Parkville, VIC, Australia.,Discipline of Optometry, University of Canberra, Canberra, ACT, Australia
| | - Shaun Frost
- CSIRO, Australian e-Health Research Centre (AEHRC), Floreat, WA, Australia
| | - Rajiv Jayasena
- CSIRO, Australian e-Health Research Centre (AEHRC), Parkville, VIC, Australia
| | - Christopher Fowler
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | | | - Haihan Jiao
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Jeremiah K H Lim
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia.,Optometry and Vision Science, College of Nursing and Health Sciences, Flinders University, Adelaide, SA, Australia
| | - Holly R Chinnery
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Laura E Downie
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
118
|
Luo J, Agboola F, Grant E, Masters CL, Albert MS, Johnson SC, McDade EM, Vöglein J, Fagan AM, Benzinger T, Massoumzadeh P, Hassenstab J, Bateman RJ, Morris JC, Perrin RJ, Chhatwal J, Jucker M, Ghetti B, Cruchaga C, Graff-Radford NR, Schofield PR, Mori H, Xiong C. Sequence of Alzheimer disease biomarker changes in cognitively normal adults: A cross-sectional study. Neurology 2020; 95:e3104-e3116. [PMID: 32873693 PMCID: PMC7734923 DOI: 10.1212/wnl.0000000000010747] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 06/12/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine the ordering of changes in Alzheimer disease (AD) biomarkers among cognitively normal individuals. METHODS Cross-sectional data, including CSF analytes, molecular imaging of cerebral fibrillar β-amyloid (Aβ) with PET using the [11C] benzothiazole tracer Pittsburgh compound B (PiB), MRI-based brain structures, and clinical/cognitive outcomes harmonized from 8 studies, collectively involving 3,284 cognitively normal individuals 18 to 101 years of age, were analyzed. The age at which each marker exhibited an accelerated change (called the change point) was estimated and compared across the markers. RESULTS Accelerated changes in CSF Aβ1-42 (Aβ42) occurred at 48.28 years of age and in Aβ42/Aβ40 ratio at 46.02 years, followed by PiB mean cortical standardized uptake value ratio (SUVR) with a change point at 54.47 years. CSF total tau (Tau) and tau phosphorylated at threonine 181 (Ptau) had a change point at ≈60 years, similar to those for MRI hippocampal volume and cortical thickness. The change point for a cognitive composite occurred at 62.41 years. The change points for CSF Aβ42 and Aβ42/Aβ40 ratio, albeit not significantly different from that for PiB SUVR, occurred significantly earlier than that for CSF Tau, Ptau, MRI markers, and the cognitive composite. Adjusted analyses confirmed that accelerated changes in CSF Tau, Ptau, MRI markers, and the cognitive composite occurred at ages not significantly different from each other. CONCLUSIONS Our findings support the hypothesized early changes of amyloid in preclinical AD and suggest that changes in neuronal injury and neurodegeneration markers occur close in time to cognitive decline.
Collapse
Affiliation(s)
- Jingqin Luo
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Folasade Agboola
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Elizabeth Grant
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Colin L Masters
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Marilyn S Albert
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Sterling C Johnson
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Eric M McDade
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Jonathan Vöglein
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Anne M Fagan
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Tammie Benzinger
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Parinaz Massoumzadeh
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Jason Hassenstab
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Randall J Bateman
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - John C Morris
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Richard J Perrin
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Jasmeer Chhatwal
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Mathias Jucker
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Bernardino Ghetti
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Carlos Cruchaga
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Neill R Graff-Radford
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Peter R Schofield
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Hiroshi Mori
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan
| | - Chengjie Xiong
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., F.A., E.G., C.X.), Knight Alzheimer Disease Research Center (F.A., E.G., A.M.F., T.B., P.M., J.H., R.J.B., J.C.M., R.J.P., C.X.), Department of Neurology (E.M.M., A.M.F., J.H., R.J.B., J.C.M., R.J.P.), Department of Radiology (T.B., P.M.), Department of Pathology (J.C.M., R.J.P.), Department of Immunology (J.C.M., R.J.P.), and Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; The Florey Institute (C.L.M.), University of Melbourne, Australia; Department of Neurology (M.S.A.), Johns Hopkins University School of Medicine, Baltimore, MD; Wisconsin Alzheimer's Institute and Alzheimer's Disease Research Center (S.C.J.), University of Wisconsin-Madison School of Medicine and Public Health; Geriatric Research Education and Clinical Center (S.C.J.), William S. Middleton Veterans Memorial Hospital, Madison, WI; German Center for Neurodegenerative Diseases (J.V.); Department of Neurology (J.V.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Neurology (J.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Hertie-Institute for Clinical Brain Research (M.J.), University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen, Germany; Department of Pathology and Laboratory Medicine (B.G.), Indiana University, Indianapolis; Department of Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; Neuroscience Research Australia (P.R.S.), Randwick; School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; and Department of Clinical Neuroscience (H.M.), Osaka City University Medical School, Abenoku, Osaka, Japan.
| |
Collapse
|
119
|
Li Z, Li K, Luo X, Zeng Q, Zhao S, Zhang B, Zhang M, Chen Y. Distinct Brain Functional Impairment Patterns Between Suspected Non-Alzheimer Disease Pathophysiology and Alzheimer's Disease: A Study Combining Static and Dynamic Functional Magnetic Resonance Imaging. Front Aging Neurosci 2020; 12:550664. [PMID: 33328953 PMCID: PMC7719833 DOI: 10.3389/fnagi.2020.550664] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 10/14/2020] [Indexed: 12/24/2022] Open
Abstract
Background: Suspected non-Alzheimer disease pathophysiology (SNAP) refers to the subjects who feature negative β-amyloid (Aβ) but positive tau or neurodegeneration biomarkers. It accounts for a quarter of the elderly population and is associated with cognitive decline. However, the underlying pathophysiology is still unclear. Methods: We included 111 non-demented subjects, then classified them into three groups using cerebrospinal fluid (CSF) Aβ 1-42 (A), phosphorylated tau 181 (T), and total tau (N). Specifically, we identified the normal control (NC; subjects with normal biomarkers, A-T-N-), SNAP (subjects with normal amyloid but abnormal tau, A-T+), and predementia Alzheimer's disease (AD; subjects with abnormal amyloid and tau, A+T+). Then, we used the static amplitude of low-frequency fluctuation (sALFF) and dynamic ALFF (dALFF) variance to reflect the intrinsic functional network strength and stability, respectively. Further, we performed a correlation analysis to explore the possible relationship between intrinsic brain activity changes and cognition. Results: SNAP showed decreased sALFF in left superior frontal gyrus (SFG) while increased sALFF in left insula as compared to NC. Regarding the dynamic metric, SNAP showed a similarly decreased dALFF in the left SFG and left paracentral lobule as compared to NC. By contrast, when compared to NC, predementia AD showed decreased sALFF in left inferior parietal gyrus (IPG) and right precuneus, while increased sALFF in the left insula, with more widely distributed decreased dALFF variance across the frontal, parietal and occipital lobe. When directly compared to SNAP, predementia AD showed decreased sALFF in left middle occipital gyrus and IPG, while showing decreased dALFF variance in the left temporal pole. Further correlation analysis showed that increased sALFF in the insula had a negative correlation with the general cognition in the SNAP group. Besides, sALFF and dALFF variance in the right precuneus negatively correlated with attention in the predementia AD group. Conclusion: SNAP and predementia AD show distinct functional impairment patterns. Specifically, SNAP has functional impairments that are confined to the frontal region, which is usually spared in early-stage AD, while predementia AD exhibits widely distributed functional damage involving the frontal, parietal and occipital cortex.
Collapse
Affiliation(s)
- Zheyu Li
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kaicheng Li
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiao Luo
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qingze Zeng
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shuai Zhao
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Baorong Zhang
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Minming Zhang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yanxing Chen
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | | |
Collapse
|
120
|
Visser PJ, Reus LM, Gobom J, Jansen I, Dicks E, Tsolaki M, Verhey FRJ, Popp J, Martinez-Lage P, Vandenberghe R, Lleó A, Molinuevo JL, Engelborghs S, Freund-Levi Y, Froelich L, Sleegers K, Dobricic V, Hong S, Lovestone S, Streffer J, Vos SJB, Bos I, Smit AB, Blennow K, Scheltens P, Teunissen CE, Bertram L, Zetterberg H, Tijms BM. Cerebrospinal fluid total tau levels indicate aberrant neuronal plasticity in Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020. [PMID: 33173883 DOI: 10.1101/2020.10.29.20211920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Alzheimer's disease (AD) is characterised by abnormal amyloid beta and tau processing. Previous studies reported that cerebrospinal fluid (CSF) total tau (t-tau) levels vary between patients. Here we show that CSF t-tau variability is associated with distinct impairments in neuronal plasticity mediated by gene repression factors SUZ12 and REST. AD individuals with abnormal t-tau levels have increased CSF concentrations of plasticity proteins regulated by SUZ12 and REST. AD individuals with normal t-tau, on the contrary, have decreased concentrations of these plasticity proteins and increased concentrations in proteins associated with blood-brain and blood CSF-barrier dysfunction. Genomic analyses suggested that t-tau levels in part depend on genes involved in gene expression. The distinct plasticity abnormalities in AD as signaled by t-tau urge the need for personalised treatment.
Collapse
|
121
|
El-Hayek YH, Wiley RE, Khoury CP, Daya RP, Ballard C, Evans AR, Karran M, Molinuevo JL, Norton M, Atri A. Tip of the Iceberg: Assessing the Global Socioeconomic Costs of Alzheimer's Disease and Related Dementias and Strategic Implications for Stakeholders. J Alzheimers Dis 2020; 70:323-341. [PMID: 31256142 PMCID: PMC6700654 DOI: 10.3233/jad-190426] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
While it is generally understood that Alzheimer’s disease (AD) and related dementias (ADRD) is one of the costliest diseases to society, there is widespread concern that researchers and policymakers are not comprehensively capturing and describing the full scope and magnitude of the socioeconomic burden of ADRD. This review aimed to 1) catalogue the different types of AD-related socioeconomic costs described in the literature; 2) assess the challenges and gaps of existing approaches to measuring these costs; and 3) analyze and discuss the implications for stakeholders including policymakers, healthcare systems, associations, advocacy groups, clinicians, and researchers looking to improve the ability to generate reliable data that can guide evidence-based decision making. A centrally emergent theme from this review is that it is challenging to gauge the true value of policies, programs, or interventions in the ADRD arena given the long-term, progressive nature of the disease, its insidious socioeconomic impact beyond the patient and the formal healthcare system, and the complexities and current deficiencies (in measures and real-world data) in accurately calculating the full costs to society. There is therefore an urgent need for all stakeholders to establish a common understanding of the challenges in evaluating the full cost of ADRD and define approaches that allow us to measure these costs more accurately, with a view to prioritizing evidence-based solutions to mitigate this looming public health crisis.
Collapse
Affiliation(s)
| | - Ryan E Wiley
- Shift Health, Toronto, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | | | | | | | | | | | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center, Barcelona, Spain.,Paqual Maragall Foundation, Barcelona, Spain
| | | | - Alireza Atri
- Banner Sun Health Research Institute, Banner Health, Sun City, AZ, USA.,Department of Neurology, Center for Brain/Mind Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
122
|
Liu Y, Li H, Wang J, Xue Q, Yang X, Kang Y, Li M, Xu J, Li G, Li C, Chang HC, Su KP, Wang F. Association of Cigarette Smoking With Cerebrospinal Fluid Biomarkers of Neurodegeneration, Neuroinflammation, and Oxidation. JAMA Netw Open 2020; 3:e2018777. [PMID: 33006621 PMCID: PMC7532384 DOI: 10.1001/jamanetworkopen.2020.18777] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 07/18/2020] [Indexed: 12/11/2022] Open
Abstract
IMPORTANCE Cigarette smoking has been associated with risk of neurodegenerative disorders, such as Alzheimer disease. The association between smoking and biomarkers of changes in human cerebrospinal fluid (CSF) is not fully understood. OBJECTIVE To investigate the association of cigarette smoking with CSF biomarkers of neurodegeneration, neuroinflammation, oxidation, and neuroprotection. DESIGN, SETTING, AND PARTICIPANTS In this case-control study of 191 adult men in China, biomarkers in the CSF of participants with and without significant cigarette exposure were examined. Participants who did not smoke and had no history of substance use disorder or dependence were assigned to the nonsmoking group. The active smoking group included participants who consumed at least 10 cigarettes per day for 1 year. Five-milliliter samples of CSF were obtained from routine lumbar puncture conducted before anterior cruciate ligament reconstruction surgery. Data collection took place from September 2014 to January 2016, and analysis took place from January to February 2016. EXPOSURES Cigarette smoking. MAIN OUTCOMES AND MEASURES CSF levels of β-amyloid 42 (Aβ42), which has diagnostic specificity for Alzheimer disease, tumor necrosis factor alpha (TNFα), brain-derived neurotrophic factor (BDNF), total superoxide dismutase (SOD), and nitric oxide synthase (NOS) were measured. Sociodemographic data and history of smoking were obtained. RESULTS Of 191 participants, 87 (45.5%) were included in the active smoking group and 104 (54.4%) in the nonsmoking group. Compared with the active smoking group, the nonsmoking group was younger (mean [SD] age, 34.4 [10.5] years vs 29.6 [9.5] years; P = .01), had more education (mean [SD] duration of education, 11.9 [3.1] years vs 13.2 [2.6] years; P = .001), and had lower body mass index (mean [SD], 25.9 [3.6] vs 24.9 [4.0]; P = .005). Comparing the nonsmoking group with the smoking group, mean (SD) CSF levels of Aβ42 (38.0 [25.9] pg/mL vs 52.8 [16.5] pg/mL; P < .001) and TNFα (23.0 [2.5] pg/mL vs 28.0 [2.0] pg/mL; P < .001) were significantly lower, while BDNF (23.1 [3.9] pg/mL vs 13.8 [2.7] pg/mL; P < .001), total SOD (15.7 [2.6] U/L vs 13.9 [2.4] U/L; P < .001), total NOS (28.3 [7.2] U/L vs 14.7 [5.6] U/L; P < .001), inducible NOS (16.0 [5.4] U/L vs 10.3 [2.7] U/L; P < .001), and constitutive NOS (12.4 [6.9] U/mL vs 4.4 [3.9] U/mL) were higher. In addition, in participants in the smoking group who were aged 40 years or older, total SOD levels were negatively correlated with Aβ42 levels (r = -0.57; P = .02). In those who smoked at least 20 cigarettes per day, TNFα levels were positively correlated with Aβ42 levels (r = 0.51; P = .006). The association of TNFα with Aβ42 production was stronger than that of total SOD with Aβ42 production (z = -4.38; P < .001). CONCLUSIONS AND RELEVANCE This case-control study found that cigarette smoking was associated with at-risk biomarkers for Alzheimer disease, as indicated by higher Aβ42 levels, excessive oxidative stress, neuroinflammation, and impaired neuroprotection found in the CSF of participants in the active smoking group.
Collapse
Affiliation(s)
- Yanlong Liu
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
- The Affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou, China
- College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hui Li
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
- Xinjiang Key Laboratory of Neurological Disorder Research, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Psychosomatic Medicine Research Division, Inner Mongolia Medical University, Huhhot, China
| | - Jian Wang
- Department of Psychology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qing Xue
- Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | | | - Yimin Kang
- Psychosomatic Medicine Research Division, Inner Mongolia Medical University, Huhhot, China
| | - Mengjie Li
- Psychosomatic Medicine Research Division, Inner Mongolia Medical University, Huhhot, China
- Sleep Medicine Center, Peking University International Hospital, Beijing, China
| | - Jinzhong Xu
- Affiliated Wenling Hospital of Wenzhou Medical University, Wenling, China
| | - Guohua Li
- Xinjiang Key Laboratory of Neurological Disorder Research, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Cunbao Li
- Psychosomatic Medicine Research Division, Inner Mongolia Medical University, Huhhot, China
| | - Hui-Chih Chang
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
- Department of Psychiatry and Mind-Body Interface Laboratory, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
| | - Kuan-Pin Su
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
- Department of Psychiatry and Mind-Body Interface Laboratory, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
- An-Nan Hospital, China Medical University, Tainan, Taiwan
| | - Fan Wang
- Xinjiang Key Laboratory of Neurological Disorder Research, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Psychosomatic Medicine Research Division, Inner Mongolia Medical University, Huhhot, China
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, China
| |
Collapse
|
123
|
Pettigrew C, Soldan A, Wang J, Wang MC, Arthur K, Moghekar A, Gottesman RF, Albert M. Association of midlife vascular risk and AD biomarkers with subsequent cognitive decline. Neurology 2020; 95:e3093-e3103. [PMID: 32989109 DOI: 10.1212/wnl.0000000000010946] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/22/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine whether vascular risk and Alzheimer disease (AD) biomarkers have independent or synergistic effects on cognitive decline and whether vascular risk is associated with the accumulation of AD pathology as measured by change in biomarkers over time. METHODS At baseline, participants (n = 168) were cognitively normal and primarily middle-aged (mean 56.4 years, SD 10.9 years) and had both vascular risk factor status and proximal CSF biomarkers available. Baseline vascular risk was quantified with a composite vascular risk score reflecting the presence or absence of hypertension, hypercholesterolemia, diabetes, current smoking, and obesity. CSF biomarkers of β-amyloid (Aβ)1-42, total tau (t-tau), and phosphorylated tau (p-tau) were used to create dichotomous high and low AD biomarker groups (based on Aβ1-42 and tau). Linear mixed-effects models were used to examine change in a cognitive composite score (mean follow-up 13.9 years) and change in CSF biomarkers (mean follow-up 4.2 years). RESULTS There was no evidence of a synergistic relationship between the vascular risk score and CSF AD biomarkers and cognitive decline. Instead, the vascular risk score (estimate -0.022, 95% confidence interval [CI] -0.043 to -0.002, p = 0.03) and AD biomarkers (estimate -0.060, 95% CI -0.096 to -0.024, p = 0.001) were independently and additively associated with cognitive decline. In addition, the vascular risk score was unrelated to levels of or rate of change in CSF Aβ1-42, t-tau, or p-tau. CONCLUSIONS The results of this observational cohort study suggest that vascular risk and biomarkers of AD pathology, when measured in midlife, act along independent pathways and underscore the importance of accounting for multiple risk factors for identifying cognitively normal individuals at the greatest risk of cognitive decline.
Collapse
Affiliation(s)
- Corinne Pettigrew
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD.
| | - Anja Soldan
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Jiangxia Wang
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Mei-Cheng Wang
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Karissa Arthur
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Abhay Moghekar
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Rebecca F Gottesman
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Marilyn Albert
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
124
|
Joannette M, Bocti C, Dupont PS, Lavallée MM, Nikelski J, Vallet GT, Chertkow H, Joubert S. Education as a Moderator of the Relationship Between Episodic Memory and Amyloid Load in Normal Aging. J Gerontol A Biol Sci Med Sci 2020; 75:1820-1826. [PMID: 31639181 PMCID: PMC7518567 DOI: 10.1093/gerona/glz235] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Indexed: 12/16/2022] Open
Abstract
The current study explored whether education, a proxy of cognitive reserve, modifies the association between episodic memory (EM) performance and βeta-amyloid load (Aβ), a biomarker of Alzheimer's disease, in a cohort of cognitively normal older adults. One hundred and four participants (mean age 73.3 years) evenly spread out in three bands of education were recruited. Participants underwent neuropsychological assessment, structural MRI as well as PET imaging to quantify Aβ load. Moderation analyses and the Johnson-Neyman technique were carried out to examine the interaction of education with Aβ load to predict EM performance. Linear regressions were then performed within each group of education to better illustrate the interaction effect (all analyses were controlled for age and sex). The interaction between education and Aβ load was significant (p < .05) for years of education, reaching a cutoff point of 13.5 years, above which the relationship between Aβ load and EM was no longer significant. Similarly, significant associations were found between Aβ and EM among participants with secondary (p < .01) and pre-university education (p < .01), but not with a university degree (p = .253). EM performance is associated with Aβ load in cognitively normal older individuals, and this relationship is moderated by educational attainment.
Collapse
Affiliation(s)
- Maude Joannette
- Département de psychologie, Université de Montréal, Montréal, Québec, Canada
- Centre de recherche de l'Institut universitaire de gériatrie de Montréal (CRIUGM), Montréal, Québec, Canada
| | - Christian Bocti
- Service de neurologie, Département de médecine, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Research Center on Aging and Memory Clinic, CIUSSS Estrie-CHUS, Sherbrooke, Québec, Canada
| | - Pénélope Sévigny Dupont
- Département de psychologie, Université de Montréal, Montréal, Québec, Canada
- Centre de recherche de l'Institut universitaire de gériatrie de Montréal (CRIUGM), Montréal, Québec, Canada
| | - Marie Maxime Lavallée
- Département de psychologie, Université de Montréal, Montréal, Québec, Canada
- Centre de recherche de l'Institut universitaire de gériatrie de Montréal (CRIUGM), Montréal, Québec, Canada
| | - Jim Nikelski
- Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec, Canada
| | - Guillaume T Vallet
- Université Clermont Auvergne, Laboratoire de Psychologie Sociale et Cognitive (CNRS, UMR6024), Clermont-Ferrand, France
| | - Howard Chertkow
- Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Sven Joubert
- Département de psychologie, Université de Montréal, Montréal, Québec, Canada
- Centre de recherche de l'Institut universitaire de gériatrie de Montréal (CRIUGM), Montréal, Québec, Canada
| |
Collapse
|
125
|
Shen X, Li J, Wang H, Li H, Huang Y, Yang Y, Tan L, Dong Q, Yu J. Plasma amyloid, tau, and neurodegeneration biomarker profiles predict Alzheimer's disease pathology and clinical progression in older adults without dementia. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2020; 12:e12104. [PMID: 33005724 PMCID: PMC7513626 DOI: 10.1002/dad2.12104] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 08/15/2020] [Accepted: 08/19/2020] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Plasma markers have been reported to be associated with brain amyloid burden, tau pathology, or neurodegeneration. We aimed to evaluate whether plasma biomarker profiles could predict Alzheimer's disease (AD) pathology and clinical progression in older adults without dementia. METHODS Cross-sectional and longitudinal data of participants enrolled in this study were from the Alzheimer's Disease Neuroimaging Initiative (ADNI). Plasma amyloid beta (Aβ)1-42/Aβ1-40 ratio was selected as the marker for amyloid pathology, p-tau181 for tau pathology, and neurofilament light for neurodegeneration. Cut-offs for these plasma markers were calculated with well-established positron emission tomography and structural imaging biomarkers as reference. Older adults without dementia were categorized into eight groups at baseline by plasma amyloid/tau/neurodegeneration (A/T/N) cut-offs. Clinical progression was analyzed using linear mixed-effects models and Cox proportional hazard models. RESULTS A total of 183 participants (97 cognitively normal [CN] subjects and 86 patients with mild cognitive impairment [MCI]; mean age 72.6 years, and 48.1% men) were included. Participants with A+ had significantly higher proportions of apolipoprotein E (APOE) gene ɛ4 carriers than those with A-. Brain atrophy was observed in all groups of CN, whereas cognition decline was obvious in the A+T+N+ group. Compared to A-T-N-, MCI patients with A+T+N+ had faster cognition worsening and faster brain atrophy. In the whole cohort, A+T+N+ and A+T+N- participants were at higher risk of clinical progression. DISCUSSION Plasma A/T/N biomarker profiles may predict AD pathology and clinical progression, indicating a potential role for plasma biomarkers in clinical trials. More research is warranted to develop a robust plasma AD framework.
Collapse
Affiliation(s)
- Xue‐Ning Shen
- Department of Neurology and Institute of NeurologyHuashan HospitalShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Jie‐Qiong Li
- Department of Neurologythe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Hui‐Fu Wang
- Department of NeurologyQingdao Municipal HospitalQingdao UniversityQingdaoChina
| | - Hong‐Qi Li
- Department of Neurology and Institute of NeurologyHuashan HospitalShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Yu‐Yuan Huang
- Department of Neurology and Institute of NeurologyHuashan HospitalShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Yu‐Xiang Yang
- Department of Neurology and Institute of NeurologyHuashan HospitalShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Lan Tan
- Department of NeurologyQingdao Municipal HospitalQingdao UniversityQingdaoChina
| | - Qiang Dong
- Department of Neurology and Institute of NeurologyHuashan HospitalShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Jin‐Tai Yu
- Department of Neurology and Institute of NeurologyHuashan HospitalShanghai Medical CollegeFudan UniversityShanghaiChina
| | | |
Collapse
|
126
|
Gao F, Shang S, Chen C, Dang L, Gao L, Wei S, Wang J, Huo K, Deng M, Wang J, Qu Q. Non-linear Relationship Between Plasma Amyloid-β 40 Level and Cognitive Decline in a Cognitively Normal Population. Front Aging Neurosci 2020; 12:557005. [PMID: 33061905 PMCID: PMC7516983 DOI: 10.3389/fnagi.2020.557005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/19/2020] [Indexed: 01/06/2023] Open
Abstract
Objectives Recent studies regarding the relationships between plasma amyloid-β (Aβ) levels and cognitive performance had inconsistent results. In this study, we aimed to characterize the relationship between cognitive decline and plasma Aβ levels in a large-sample cognitively normal population. Methods This population-based, prospective cohort study included 1,240 participants with normal cognition. The Mini-Mental State Examination (MMSE) was used to assess cognitive function at baseline and 2 years later. Restricted cubic splines, multivariate logistic regression, and multivariate linear regression models were used to evaluate the type of relationship between cognitive decline during the 2-year follow-up period and plasma Aβ levels (Aβ40, Aβ42, and Aβ42/40). Results Participants with moderate Aβ40 levels had the highest risk of cognitive decline during a 2-year follow-up relative to individuals with low Aβ40 [odds ratio (OR): 0.60, 95% confidence interval (CI): 0.45–0.81, p < 0.001] or high Aβ40 (OR: 0.65, 95% CI: 0.49–0.87, p = 0.004) levels. The association between Aβ40 and cognitive decline did not depend on sex, education level, or APOE ε4 status. There was an interaction found between age (≤ 65 and > 65 years) and Aβ40 (p for interaction = 0.021). In individuals older than 65 years, there was a positive linear relationship between plasma Aβ40 and cognitive decline (OR: 1.02, 95% CI: 1.00–1.04, p = 0.027). For participants ≤ 65 years old, the lower Aβ40 and higher Aβ40 groups had a lower risk of cognitive decline than the medium Aβ40 group (OR: 0.69, 95% CI: 0.50–0.94, p = 0.02; OR: 0.63, 95% CI: 0.45–0.86, p = 0.004). None of relationship between plasma Aβ42, Aβ42/40 and cognitive decline was found during a 2-year follow-up. Conclusion The relationship between plasma Aβ40 and cognitive decline was not linear, but an inverted-U shape in a cognitively normal population. The underlying mechanism requires further investigation.
Collapse
Affiliation(s)
- Fan Gao
- Clinical Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Suhang Shang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chen Chen
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liangjun Dang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ling Gao
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shan Wei
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Wang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kang Huo
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meiying Deng
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jingyi Wang
- Department of Neurology, Huxian Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Qiumin Qu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
127
|
Significance of Blood and Cerebrospinal Fluid Biomarkers for Alzheimer's Disease: Sensitivity, Specificity and Potential for Clinical Use. J Pers Med 2020; 10:jpm10030116. [PMID: 32911755 PMCID: PMC7565390 DOI: 10.3390/jpm10030116] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/21/2020] [Accepted: 09/01/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia, affecting more than 5 million Americans, with steadily increasing mortality and incredible socio-economic burden. Not only have therapeutic efforts so far failed to reach significant efficacy, but the real pathogenesis of the disease is still obscure. The current theories are based on pathological findings of amyloid plaques and tau neurofibrillary tangles that accumulate in the brain parenchyma of affected patients. These findings have defined, together with the extensive neurodegeneration, the diagnostic criteria of the disease. The ability to detect changes in the levels of amyloid and tau in cerebrospinal fluid (CSF) first, and more recently in blood, has allowed us to use these biomarkers for the specific in-vivo diagnosis of AD in humans. Furthermore, other pathological elements of AD, such as the loss of neurons, inflammation and metabolic derangement, have translated to the definition of other CSF and blood biomarkers, which are not specific of the disease but, when combined with amyloid and tau, correlate with the progression from mild cognitive impairment to AD dementia, or identify patients who will develop AD pathology. In this review, we discuss the role of current and hypothetical biomarkers of Alzheimer's disease, their specificity, and the caveats of current high-sensitivity platforms for their peripheral detection.
Collapse
|
128
|
Grey zone amyloid burden affects memory function: the SCIENCe project. Eur J Nucl Med Mol Imaging 2020; 48:747-756. [PMID: 32888039 PMCID: PMC8036199 DOI: 10.1007/s00259-020-05012-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/20/2020] [Indexed: 12/24/2022]
Abstract
Purpose To determine thresholds for amyloid beta pathology and evaluate associations with longitudinal memory performance with the aim to identify a grey zone of early amyloid beta accumulation and investigate its clinical relevance. Methods We included 162 cognitively normal participants with subjective cognitive decline from the SCIENCe cohort (64 ± 8 years, 38% F, MMSE 29 ± 1). Each underwent a dynamic [18F] florbetapir PET scan, a T1-weighted MRI scan and longitudinal memory assessments (RAVLT delayed recall, n = 655 examinations). PET scans were visually assessed as amyloid positive/negative. Additionally, we calculated the mean binding potential (BPND) and standardized uptake value ratio (SUVr50–70) for an a priori defined composite region of interest. We determined six amyloid positivity thresholds using various data-driven methods (resulting thresholds: BPND 0.19/0.23/0.29; SUVr 1.28/1.34/1.43). We used Cohen’s kappa to analyse concordance between thresholds and visual assessment. Next, we used quantiles to divide the sample into two to five subgroups of equal numbers (median, tertiles, quartiles, quintiles), and operationalized a grey zone as the range between the thresholds (0.19–0.29 BPND/1.28–1.43 SUVr). We used linear mixed models to determine associations between thresholds and memory slope. Results As determined by visual assessment, 24% of 162 individuals were amyloid positive. Concordance with visual assessment was comparable but slightly higher for BPND thresholds (range kappa 0.65–0.70 versus 0.60–0.63). All thresholds predicted memory decline (range beta − 0.29 to − 0.21, all p < 0.05). Analyses in subgroups showed memory slopes gradually became steeper with higher amyloid load (all p for trend < 0.05). Participants with a low amyloid burden benefited from a practice effect (i.e. increase in memory), whilst high amyloid burden was associated with memory decline. Memory slopes of individuals in the grey zone were intermediate. Conclusion We provide evidence that not only high but also grey zone amyloid burden subtly impacts memory function. Therefore, in case a binary classification is required, we suggest using a relatively low threshold which includes grey zone amyloid pathology.
Collapse
|
129
|
Yang HS, Chhatwal JP, Xu J, White CC, Hanseeuw B, Rabin JS, Papp KV, Buckley RF, Schultz AP, Properzi MJ, Gatchel JR, Amariglio RE, Donovan NJ, Mormino EC, Hedden T, Marshall GA, Rentz DM, Johnson KA, De Jager PL, Sperling RA. An UNC5C Allele Predicts Cognitive Decline and Hippocampal Atrophy in Clinically Normal Older Adults. J Alzheimers Dis 2020; 68:1161-1170. [PMID: 30883345 DOI: 10.3233/jad-180788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The UNC5C rs3846455G allele has been linked to poor cognitive resilience against age-related neuropathologies, but this association remains to be replicated, and the allele's effect on hippocampal neurodegeneration needs to be examined. OBJECTIVE To further validate the association between rs3846455G and faster cognitive decline, especially among cognitively normal older adults, and to assess whether rs3846455G predicts accelerated hippocampal volume loss in older adults. METHODS We assessed participants in the Harvard Aging Brain Study (HABS), a longitudinal cohort study of older adults who were clinically normal at baseline. To avoid bias from population admixture, analyses were limited to participants of European descent with longitudinal neuroimaging data (n = 174). Linear mixed effect models were used to examine the effect of rs3846455G on longitudinal change of the Preclinical Alzheimer Cognitive Composite (PACC) and MRI-measured bilateral hippocampal volume, adjusting for baseline amyloid-β (Aβ) measured by the cortical Pittsburgh Compound B PET distributed volume ratio. We also tested whether hippocampal atrophy mediates the association between rs3846455G and greater PACC decline through a mediation analysis. RESULTS rs3846455G was associated with greater PACC decline (β= -0.087/year, 95% CI -0.169 to -0.005, p = 0.039) after controlling for baseline Aβ. Further, rs3846455G predicted accelerated hippocampal atrophy after controlling for baseline Aβ (β= -57.3 mm3/year, 95% CI -102.8 to -11.9, p = 0.014). The association between rs3846455G and greater PACC decline was partially mediated by accelerated hippocampal atrophy (mediated effect (relative scale) = -0.014, 95% CI -0.032 to -6.0×10-4, p = 0.039). CONCLUSION UNC5C rs3846455G predicts greater cognitive decline and accelerated hippocampal atrophy in clinically normal older adults.
Collapse
Affiliation(s)
- Hyun-Sik Yang
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Cell Circuits Program, Broad Institute of MIT and Harvard, Cambridge, MAs, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Jishu Xu
- Cell Circuits Program, Broad Institute of MIT and Harvard, Cambridge, MAs, USA
| | - Charles C White
- Cell Circuits Program, Broad Institute of MIT and Harvard, Cambridge, MAs, USA
| | - Bernard Hanseeuw
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Department of Neurology, Cliniques Universitaires Saint-Luc, Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
| | - Jennifer S Rabin
- Harvard Medical School, Boston, MA, USA.,Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Kathryn V Papp
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Rachel F Buckley
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Florey Institutes of Neuroscience and Mental Health, Melbourne, VIC, Australia.,Melbourne School of Psychological Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Michael J Properzi
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Jennifer R Gatchel
- Harvard Medical School, Boston, MA, USA.,Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.,Division of Geriatric Psychiatry, McLean Hospital, Belmont, MA, USA.,Gerontology Research Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Rebecca E Amariglio
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Nancy J Donovan
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.,Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, USA
| | - Elizabeth C Mormino
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Trey Hedden
- Harvard Medical School, Boston, MA, USA.,Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Gad A Marshall
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Dorene M Rentz
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Philip L De Jager
- Cell Circuits Program, Broad Institute of MIT and Harvard, Cambridge, MAs, USA.,Department of Neurology, Center for Translational & Computational Neuroimmunology, Columbia University Medical Center, New York, NY, USA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
130
|
Lozupone M, Solfrizzi V, D'Urso F, Di Gioia I, Sardone R, Dibello V, Stallone R, Liguori A, Ciritella C, Daniele A, Bellomo A, Seripa D, Panza F. Anti-amyloid-β protein agents for the treatment of Alzheimer's disease: an update on emerging drugs. Expert Opin Emerg Drugs 2020; 25:319-335. [PMID: 32772738 DOI: 10.1080/14728214.2020.1808621] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Currently available Alzheimer's disease (AD) therapeutics are only symptomatic, targeting cholinergic and glutamatergic neurotransmissions. Several putative disease-modifying drugs in late-stage clinical development target amyloid-β (Aβ) peptide and tau protein, the principal neurophatological hallmarks of the disease. AREAS COVERED Phase III randomized clinical trials of anti-Aβ drugs for AD treatment were searched in US and EU clinical trial registries and principal biomedical databases until May 2020. EXPERT OPINION At present, compounds in Phase III clinical development for AD include four anti-Ab monoclonal antibodies (solanezumab, gantenerumab, aducanumab, BAN2401), the combination of cromolyn sodium and ibuprofen (ALZT-OP1), and two small molecules (levetiracetam, GV-971). These drugs are mainly being tested in subjects during early AD phases or at preclinical stage of familial AD or even in asymptomatic subjects at high risk of developing AD. The actual results support the hypothesis that elevated Aβ represents an early stage in the AD continuum and demonstrate the feasibility of enrolling these high-risk participants in secondary prevention trials to slow cognitive decline during the AD preclinical stages. However, a series of clinical failures may question further development of Aβ-targeting drugs and the findings from current ongoing Phase III trials will hopefully give light to this critical issue.
Collapse
Affiliation(s)
- Madia Lozupone
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro , Bari, Italy
| | - Vincenzo Solfrizzi
- "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari "Aldo Moro" , Bari, Italy
| | - Francesca D'Urso
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia , Foggia, Italy
| | - Ilaria Di Gioia
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia , Foggia, Italy
| | - Rodolfo Sardone
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy
| | - Vittorio Dibello
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy.,Department of Orofacial Pain and Dysfunction, Academic Centre of Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam , The Netherlands
| | - Roberta Stallone
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy
| | - Angelo Liguori
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy
| | - Chiara Ciritella
- Physical and Rehabilitation Medicine Department, University of Foggia , Foggia, Italy
| | - Antonio Daniele
- Institute of Neurology, Catholic University of Sacred Heart , Rome, Italy.,Institute of Neurology, Fondazione Policlinico Universitario A. Gemelli IRCCS , Rome, Italy
| | - Antonello Bellomo
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia , Foggia, Italy
| | - Davide Seripa
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS Casa Sollievo Della Sofferenza , Foggia, Italy.,Hematology and Stem Cell Transplant Unit, Vito Fazzi Hospital, ASL Lecce , Lecce, Italy
| | - Francesco Panza
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy
| |
Collapse
|
131
|
Millar PR, Ances BM, Gordon BA, Benzinger TLS, Fagan AM, Morris JC, Balota DA. Evaluating resting-state BOLD variability in relation to biomarkers of preclinical Alzheimer's disease. Neurobiol Aging 2020; 96:233-245. [PMID: 33039901 DOI: 10.1016/j.neurobiolaging.2020.08.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/21/2020] [Accepted: 08/10/2020] [Indexed: 02/09/2023]
Abstract
Recent functional magnetic resonance imaging studies have demonstrated that moment-to-moment variability in the blood oxygen level-dependent (BOLD) signal is related to age differences, cognition, and symptomatic Alzheimer's disease (AD). However, no studies have examined BOLD variability in the context of preclinical AD. We tested relationships between resting-state BOLD variability and biomarkers of amyloidosis, tauopathy, and neurodegeneration in a large (N = 321), well-characterized sample of cognitively normal adults (age = 39-93), using multivariate machine learning techniques. Furthermore, we controlled for cardiovascular health factors, which may contaminate resting-state BOLD variability estimates. BOLD variability, particularly in the default mode network, was related to cerebrospinal fluid (CSF) amyloid-β42 but was not related to CSF phosphorylated tau-181. Furthermore, BOLD variability estimates were also related to markers of neurodegeneration, including CSF neurofilament light protein, hippocampal volume, and a cortical thickness composite. Notably, relationships with hippocampal volume and cortical thickness survived correction for cardiovascular health and also contributed to age-related differences in BOLD variability. Thus, BOLD variability may be sensitive to preclinical pathology, including amyloidosis and neurodegeneration in AD-sensitive areas.
Collapse
Affiliation(s)
- Peter R Millar
- Department of Psychological & Brain Sciences, St. Louis, MO, USA; Department of Neurology, St. Louis, MO, USA.
| | - Beau M Ances
- Department of Neurology, St. Louis, MO, USA; Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Brian A Gordon
- Department of Psychological & Brain Sciences, St. Louis, MO, USA; Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | | | | | | | - David A Balota
- Department of Psychological & Brain Sciences, St. Louis, MO, USA; Department of Neurology, St. Louis, MO, USA
| |
Collapse
|
132
|
Spencer BE, Jennings RG, Brewer JB. Combined Biomarker Prognosis of Mild Cognitive Impairment: An 11-Year Follow-Up Study in the Alzheimer's Disease Neuroimaging Initiative. J Alzheimers Dis 2020; 68:1549-1559. [PMID: 30958366 DOI: 10.3233/jad-181243] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Biomarkers may soon be used to predict decline in older individuals. Extended follow-up studies are needed to determine the stability of such biomarker-based predictions. OBJECTIVE To examine the long-term performance of baseline cognitive, neuroimaging, and cerebrospinal fluid (CSF) biomarker-assisted prognosis in patients with mild cognitive impairment. METHODS Established, biomarker-defined, cohorts of subjects with mild cognitive impairment were examined for progression to dementia. Subjects with a baseline volumetric MRI, lumbar puncture, and Rey Auditory Verbal Learning Test were included. Dementia-free survival time in each biomarker-defined risk group was determined with Kaplan-Meier survival curves. The influence of each risk factor or combination of factors on dementia-free survival was examined with Cox proportional hazard analyses. RESULTS 185 subjects were followed longitudinally for a mean (SD) 4.3 (2.8) years. 59% of participants converted within the follow-up period and the median dementia-free survival time was 2.8 years. Each individual risk factor predicted conversion to dementia (HR 1.9-3.7). The joint presence of any two risk factors increased risk for conversion (HR 7.1-11.0), with the presence of medial temporal atrophy and memory impairment showing the greatest risk for decline. Concordant atrophy, memory impairment, and abnormal CSF amyloid and tau was associated with the highest risk for conversion (HR 15.1). The presence of medial temporal atrophy was associated with the shortest dementia-free survival time, both alone and in combination with memory impairment, abnormal CSF amyloid and tau, or both. CONCLUSION These results suggest that baseline biomarker-assisted predictions of decline to dementia are stable over the long term, and that combinations of complementary biomarkers can improve the accuracy of these predictions.
Collapse
Affiliation(s)
- Barbara E Spencer
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Robin G Jennings
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - James B Brewer
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA.,Department of Radiology, University of California, San Diego, La Jolla, CA, USA
| | | |
Collapse
|
133
|
Uddin MS, Kabir MT, Rahman MS, Behl T, Jeandet P, Ashraf GM, Najda A, Bin-Jumah MN, El-Seedi HR, Abdel-Daim MM. Revisiting the Amyloid Cascade Hypothesis: From Anti-Aβ Therapeutics to Auspicious New Ways for Alzheimer's Disease. Int J Mol Sci 2020; 21:5858. [PMID: 32824102 PMCID: PMC7461598 DOI: 10.3390/ijms21165858] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/03/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder related to age, characterized by the cerebral deposition of fibrils, which are made from the amyloid-β (Aβ), a peptide of 40-42 amino acids. The conversion of Aβ into neurotoxic oligomeric, fibrillar, and protofibrillar assemblies is supposed to be the main pathological event in AD. After Aβ accumulation, the clinical symptoms fall out predominantly due to the deficient brain clearance of the peptide. For several years, researchers have attempted to decline the Aβ monomer, oligomer, and aggregate levels, as well as plaques, employing agents that facilitate the reduction of Aβ and antagonize Aβ aggregation, or raise Aβ clearance from brain. Unluckily, broad clinical trials with mild to moderate AD participants have shown that these approaches were unsuccessful. Several clinical trials are running involving patients whose disease is at an early stage, but the preliminary outcomes are not clinically impressive. Many studies have been conducted against oligomers of Aβ which are the utmost neurotoxic molecular species. Trials with monoclonal antibodies directed against Aβ oligomers have exhibited exciting findings. Nevertheless, Aβ oligomers maintain equivalent states in both monomeric and aggregation forms; so, previously administered drugs that precisely decrease Aβ monomer or Aβ plaques ought to have displayed valuable clinical benefits. In this article, Aβ-based therapeutic strategies are discussed and several promising new ways to fight against AD are appraised.
Collapse
Affiliation(s)
- Md. Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka 1213, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka 1207, Bangladesh
| | - Md. Tanvir Kabir
- Department of Pharmacy, BRAC University, Dhaka 1212, Bangladesh;
| | - Md. Sohanur Rahman
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi 6205, Bangladesh;
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Philippe Jeandet
- Research Unit, Induced Resistance and Plant Bioprotection, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, University of Reims Champagne-Ardenne, PO Box 1039, 51687 Reims CEDEX 2, France;
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Agnieszka Najda
- Laboratory of Quality of Vegetables and Medicinal Plants, Department of Vegetable Crops and Medicinal Plants, University of Life Sciences in Lublin, 15 Akademicka Street, 20-950 Lublin, Poland;
| | - May N. Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia;
| | - Hesham R. El-Seedi
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China;
- Pharmacognosy Group, Department of Pharmaceutical Biosciences, Uppsala University, SE-751 23 Uppsala, Sweden
- Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Koom 32512, Egypt
| | - Mohamed M. Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
134
|
Aydemir D, Ulusu NN. Importance of the serum biochemical parameters as potential biomarkers for rapid diagnosis and evaluating preclinical stage of ALS. Med Hypotheses 2020; 141:109736. [DOI: 10.1016/j.mehy.2020.109736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/05/2020] [Accepted: 04/09/2020] [Indexed: 02/07/2023]
|
135
|
It's complicated: The relationship between sleep and Alzheimer's disease in humans. Neurobiol Dis 2020; 144:105031. [PMID: 32738506 DOI: 10.1016/j.nbd.2020.105031] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/21/2020] [Accepted: 07/26/2020] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by an asymptomatic period of amyloid-β (Aβ) deposition as insoluble extracellular plaque, intracellular tau aggregation, neuronal and synaptic loss, and subsequent cognitive dysfunction and dementia. A growing public health crisis, the worldwide prevalence of AD is expected to rise from 46.8 million individuals affected in 2015 to 131.5 million in 2050. Sleep disturbances have been associated with increased future risk of AD. A bi-directional relationship is hypothesized between sleep and AD with sleep disturbances as either markers for AD pathology and/or a mechanism mediating increased risk of AD. In this review, the evidence in humans supporting this complex relationship between sleep and AD will be discussed as well as the therapeutic potential and challenges of treating sleep disturbances to prevent or delay the onset of AD.
Collapse
|
136
|
Jiao F, Yi F, Wang Y, Zhang S, Guo Y, Du W, Gao Y, Ren J, Zhang H, Liu L, Song H, Wang L. The Validation of Multifactor Model of Plasma Aβ 42 and Total-Tau in Combination With MoCA for Diagnosing Probable Alzheimer Disease. Front Aging Neurosci 2020; 12:212. [PMID: 32792940 PMCID: PMC7385244 DOI: 10.3389/fnagi.2020.00212] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/16/2020] [Indexed: 01/21/2023] Open
Abstract
Alzheimer disease (AD) has an insidious onset and heterogeneous clinical symptoms. The well-accepted biomarkers for clinical diagnosis of AD include β-amyloid (Aβ) deposition and pathologic tau level within cerebral spinal fluid (CSF) and imaging AD pathology such as positive emission tomography (PET) imaging of the amyloid-binding agent Pittsburgh compound B (PET-PiB). However, the high expense and invasive nature of these methods highly limit their wide usage in clinic practice. Therefore, it is imperious to develop less expensive and invasive methods, and plasma biomarkers are the premium targets. In the current study, we utilized a single-blind comparison method; all the probable AD cases met the core clinical National Institute on Aging and Alzheimer’s Association (NIA-AA) criteria and validated by PET-PiB. We used ultrasensitive immunomagnetic reduction (IMR) assays to measure plasma Aβ42 and total-tau (t-tau) levels, in combination with different variables including Aβ42 × t-tau value, Montreal Cognitive Assessment (MoCA), and Mini Mental State Examination (MMSE). We used logistic regression to analyze the effect of all these variables in the algorism. Our results showed that (1) plasma Aβ42 and t-tau are efficient biomarkers for AD diagnosis using IMR platform, whereas Aβ42 × t-tau value is more efficient for discriminating control and AD; (2) in the control group, Aβ42 level and age demonstrated strong negative correlation; Aβ42 × t-tau value and age demonstrated significant negative correlation; (3) in the AD group, t-tau level and MMSE score demonstrated strong negative correlation; (4) using the model that Aβ42, Aβ42 × t-tau, and MoCA as the variable to generate receiver operating characteristic (ROC) curve, cutoff value = 0.48, sensitivity = 0.973, specificity = 0.982, area under the curve (AUC) = 0.986, offered better categorical efficacy, sensitivity, specificity, and AUC. The multifactor model of plasma Aβ42 and t-tau in combination with MoCA can be a viable model separate health and AD subjects in clinical practice.
Collapse
Affiliation(s)
- Fubin Jiao
- Medical School of Chinese People's Liberation Army, Beijing, China.,Department of Neurology, The 2nd Medical Center, National Clinical Research Center for Geriatric Disease, Chinese People's Liberation Army General Hospital, Beijing, China.,Health Service Department of the Guard Bureau of the Joint Staff Department, Joint Staff of the Central Military Commission of Chinese PLA, Beijing, China
| | - Fang Yi
- Department of Neurology, The 2nd Medical Center, National Clinical Research Center for Geriatric Disease, Chinese People's Liberation Army General Hospital, Beijing, China.,Department of Neurology, Lishilu Outpatient, Jingzhong Medical District, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yuanyuan Wang
- Department of Neurology, The 2nd Medical Center, National Clinical Research Center for Geriatric Disease, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Shouzi Zhang
- The Psycho Department of Beijing Geriatric Hospital, Beijing, China
| | - Yanjun Guo
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wenjin Du
- Department of Neurology, Air Force Medical Center, Chinese People's Liberation Army, Beijing, China
| | - Ya Gao
- Department of Geriatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jingjing Ren
- National Engineering Research Center for Protein Drugs, Beijing, China
| | - Haifeng Zhang
- Health Service Department of the Guard Bureau of the Joint Staff Department, Joint Staff of the Central Military Commission of Chinese PLA, Beijing, China
| | - Lixin Liu
- The Psycho Department of Beijing Geriatric Hospital, Beijing, China
| | - Haifeng Song
- National Engineering Research Center for Protein Drugs, Beijing, China
| | - Luning Wang
- Medical School of Chinese People's Liberation Army, Beijing, China.,Department of Neurology, The 2nd Medical Center, National Clinical Research Center for Geriatric Disease, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
137
|
Guo S, Xiao B, Wu C. Identifying subtypes of mild cognitive impairment from healthy aging based on multiple cortical features combined with volumetric measurements of the hippocampal subfields. Quant Imaging Med Surg 2020; 10:1477-1489. [PMID: 32676366 DOI: 10.21037/qims-19-872] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Background Mild cognitive impairment (MCI) is subtle cognitive decline with an estimated 10-15% yearly conversion rate toward Alzheimer's disease (AD). It remains unexplored in brain cortical association areas in different lobes and its changes with progression and conversion of MCI. Methods Brain structural magnetic resonance (MR) images were collected from 102 stable MCI (sMCI) patients. One hundred eleven were converted MCI (cMCI) patients, and 109 were normal control (NC). The cortical surface features and volumes of subcortical hippocampal subfields were calculated using the FreeSurfer software, followed by an analysis of variance (ANOVA) model, to reveal the differences between the NC-sMCI, NC-cMCI, and sMCI-cMCI groups. Afterward, the support vector machine-recursive feature elimination (SVM-RFE) method was applied to determine the differences between the groups. Results The experimental results showed that there were progressive degradations in either range or degree of the brain structure from NC to sMCI, and then to cMCI. The SVM classifier obtained accuracies with 64.62%, 78.96%, and 70.33% in the sMCI-NC, cMCI-NC, and cMCI-sMCI groups, respectively, using the volumes of hippocampal subfields independently. The combination of the volumes from the hippocampal subfields and cortical measurements could significantly increase the performance to 71.86%, 84.64%, and 76.86% for the sMCI-NC, cMCI-NC, and cMCI-sMCI classifications, respectively. Also, the brain regions corresponding to the dominant features with strong discriminative power were widely located in the temporal, frontal, parietal, olfactory cortexes, and most of the hippocampal subfields, which were associated with cognitive decline, memory impairment, spatial navigation, and attention control. Conclusions The combination of cortical features with the volumes of hippocampal subfields could supply critical information for MCI detection and its conversion.
Collapse
Affiliation(s)
- Shengwen Guo
- Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, Guangzhou, China
| | - Benheng Xiao
- Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, Guangzhou, China
| | - Congling Wu
- Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, Guangzhou, China
| | | |
Collapse
|
138
|
Ebenau JL, Timmers T, Wesselman LMP, Verberk IMW, Verfaillie SCJ, Slot RER, van Harten AC, Teunissen CE, Barkhof F, van den Bosch KA, van Leeuwenstijn M, Tomassen J, Braber AD, Visser PJ, Prins ND, Sikkes SAM, Scheltens P, van Berckel BNM, van der Flier WM. ATN classification and clinical progression in subjective cognitive decline: The SCIENCe project. Neurology 2020; 95:e46-e58. [PMID: 32522798 PMCID: PMC7371376 DOI: 10.1212/wnl.0000000000009724] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 12/12/2019] [Indexed: 02/06/2023] Open
Abstract
Objective To investigate the relationship between the ATN classification system (amyloid, tau, neurodegeneration) and risk of dementia and cognitive decline in individuals with subjective cognitive decline (SCD). Methods We classified 693 participants with SCD (60 ± 9 years, 41% women, Mini-Mental State Examination score 28 ± 2) from the Amsterdam Dementia Cohort and Subjective Cognitive Impairment Cohort (SCIENCe) project according to the ATN model, as determined by amyloid PET or CSF β-amyloid (A), CSF p-tau (T), and MRI-based medial temporal lobe atrophy (N). All underwent extensive neuropsychological assessment. For 342 participants, follow-up was available (3 ± 2 years). As a control population, we included 124 participants without SCD. Results Fifty-six (n = 385) participants had normal Alzheimer disease (AD) biomarkers (A–T–N–), 27% (n = 186) had non-AD pathologic change (A–T–N+, A–T+N–, A–T+N+), 18% (n = 122) fell within the Alzheimer continuum (A+T–N–, A+T–N+, A+T+N–, A+T+N+). ATN profiles were unevenly distributed, with A–T+N+, A+T–N+, and A+T+N+ containing very few participants. Cox regression showed that compared to A–T–N–, participants in A+ profiles had a higher risk of dementia with a dose–response pattern for number of biomarkers affected. Linear mixed models showed participants in A+ profiles showed a steeper decline on tests addressing memory, attention, language, and executive functions. In the control group, there was no association between ATN and cognition. Conclusions Among individuals presenting with SCD at a memory clinic, those with a biomarker profile A–T+N+, A+T–N–, A+T+N–, and A+T+N+ were at increased risk of dementia, and showed steeper cognitive decline compared to A–T–N– individuals. These results suggest a future where biomarker results could be used for individualized risk profiling in cognitively normal individuals presenting at a memory clinic.
Collapse
Affiliation(s)
- Jarith L Ebenau
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden.
| | - Tessa Timmers
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Linda M P Wesselman
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Inge M W Verberk
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Sander C J Verfaillie
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Rosalinde E R Slot
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Argonde C van Harten
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Charlotte E Teunissen
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Frederik Barkhof
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Karlijn A van den Bosch
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Mardou van Leeuwenstijn
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Jori Tomassen
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Anouk den Braber
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Pieter Jelle Visser
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Niels D Prins
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Sietske A M Sikkes
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Philip Scheltens
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Bart N M van Berckel
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Wiesje M van der Flier
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| |
Collapse
|
139
|
Dicks E, van der Flier WM, Scheltens P, Barkhof F, Tijms BM. Single-subject gray matter networks predict future cortical atrophy in preclinical Alzheimer's disease. Neurobiol Aging 2020; 94:71-80. [PMID: 32585492 DOI: 10.1016/j.neurobiolaging.2020.05.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 05/08/2020] [Accepted: 05/10/2020] [Indexed: 02/07/2023]
Abstract
The development of preventive strategies in early-stage Alzheimer's disease (AD) requires measures that can predict future brain atrophy. Gray matter network measures are related to amyloid burden in cognitively normal older individuals and predict clinical progression in preclinical AD. Here, we show that within individuals with preclinical AD, gray matter network measures predict hippocampal atrophy rates, whereas other AD biomarkers (total gray matter volume, cerebrospinal fluid total tau, and Mini-Mental State Examination) do not. Furthermore, in brain areas where amyloid is known to start aggregating (i.e. anterior cingulate and precuneus), disrupted network measures predict faster atrophy in other distant areas, mostly involving temporal regions, which are associated with AD. When repeating analyses in age-matched, cognitively unimpaired individuals without amyloid or tau pathology, we did not find any associations between network measures and hippocampal atrophy, suggesting that the associations are specific for preclinical AD. Our findings suggest that disrupted gray matter networks may indicate a treatment opportunity in preclinical AD individuals but before the onset of irreversible atrophy and cognitive impairment.
Collapse
Affiliation(s)
- Ellen Dicks
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands.
| | - Wiesje M van der Flier
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; Department of Epidemiology and Biostatistics, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Philip Scheltens
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; Centre for Medical Image Computing, Medical Physics and Biomedical Engineering, UCL, London, United Kingdom
| | - Betty M Tijms
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | | |
Collapse
|
140
|
Nguyen TT, Ta QTH, Nguyen TKO, Nguyen TTD, Vo VG. Role of Body-Fluid Biomarkers in Alzheimer's Disease Diagnosis. Diagnostics (Basel) 2020; 10:diagnostics10050326. [PMID: 32443860 PMCID: PMC7277970 DOI: 10.3390/diagnostics10050326] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/02/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD) is a complex neurodegenerative disease that requires extremely specific biomarkers for its diagnosis. For current diagnostics capable of identifying AD, the development and validation of early stage biomarkers is a top research priority. Body-fluid biomarkers might closely reflect synaptic dysfunction in the brain and, thereby, could contribute to improving diagnostic accuracy and monitoring disease progression, and serve as markers for assessing the response to disease-modifying therapies at early onset. Here, we highlight current advances in the research on the capabilities of body-fluid biomarkers and their role in AD pathology. Then, we describe and discuss current applications of the potential biomarkers in clinical diagnostics in AD.
Collapse
Affiliation(s)
- Thuy Trang Nguyen
- Faculty of Pharmacy, Ho Chi Minh City University of Technology (HUTECH), Ho Chi Minh City 700000, Vietnam;
| | - Qui Thanh Hoai Ta
- Institute of Research and Development, Duy Tan University, Danang 550000, Vietnam;
| | - Thi Kim Oanh Nguyen
- Faculty of Food Science and Technology, Ho Chi Minh City University of Food Industry, Ho Chi Minh City 700000, Vietnam;
| | - Thi Thuy Dung Nguyen
- Faculty of Environmental and Food Engineering, Nguyen Tat Thanh University, Ho Chi Minh City 70000, Vietnam
- Correspondence: (T.T.D.N.); (V.G.V.)
| | - Van Giau Vo
- Department of Industrial and Environmental Engineering, Graduate School of Environment, Gachon University, 1342 Sungnam-daero, Sujung-gu, Seongnam-si, Gyeonggi-do 461-701, Korea
- Department of BionanoTechnology, Gachon University, 1342 Sungnam-daero, Sujung-gu, Seongnam-si, Gyeonggi-do 461-701, Korea
- Correspondence: (T.T.D.N.); (V.G.V.)
| |
Collapse
|
141
|
Crous-Bou M, Gascon M, Gispert JD, Cirach M, Sánchez-Benavides G, Falcon C, Arenaza-Urquijo EM, Gotsens X, Fauria K, Sunyer J, Nieuwenhuijsen MJ, Luis Molinuevo J. Impact of urban environmental exposures on cognitive performance and brain structure of healthy individuals at risk for Alzheimer's dementia. ENVIRONMENT INTERNATIONAL 2020; 138:105546. [PMID: 32151419 DOI: 10.1016/j.envint.2020.105546] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 05/07/2023]
Abstract
BACKGROUND Air quality might contribute to incidence of dementia-related disorders, including Alzheimer's dementia (AD). The aim of our study is to evaluate the effect of urban environmental exposures (including exposure to air pollution, noise and green space) on cognitive performance and brain structure of cognitively unimpaired individuals at risk for AD. PARTICIPANTS AND METHODS The ALFA (ALzheimer and FAmilies) study is a prospective cohort of middle-age, cognitively unimpaired subjects, many of them offspring of AD patients. Cognitive performance was measured by the administration of episodic memory and executive function tests (N = 958). Structural brain imaging was performed in a subsample of participants to obtain morphological information of brain areas, specially focused on cortical thickness, known to be affected by AD (N = 228). Land Use Regression models were used to estimate residential exposure to air pollutants. The daily average noise level at the street nearest to each participant's residential address was obtained from noise maps. For each participant residential green exposure indicators, such as surrounding greenness or amount of green, were generated. General linear models were conducted to assess the association between environmental exposures, cognitive performance and brain structure in a cross-sectional analysis. RESULTS No significant associations were observed between urban environmental exposures and the cognitive composite (p > 0.1). Higher exposure to air pollutants, but not noise, was associated with lower cortical thickness in brain regions known to be affected by AD, especially NO2 (β = -16.4; p = 0.05) and PM10 (β = -5.34; p = 0.05). On the other hand, increasing greenness indicators was associated with greater thickness in these same areas (β = 0.08; p = 0.03). CONCLUSION In cognitively unimpaired adults with increased risk for AD, increased exposure to air pollution was suggested to be associated with greater global atrophy and reduced volume and thickness in specific brain areas known to be affected in AD, thus suggesting a potential link between environmental exposures and cerebral vulnerability to AD. Although more research in the field is needed, air pollution reduction is crucial for decreasing the burden of age-related disorders.
Collapse
Affiliation(s)
- Marta Crous-Bou
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Mireia Gascon
- Universitat Pompeu Fabra (UPF), Barcelona, Spain; ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain
| | - Marta Cirach
- Universitat Pompeu Fabra (UPF), Barcelona, Spain; ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Gonzalo Sánchez-Benavides
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Carles Falcon
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; CIBER Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain
| | - Eider M Arenaza-Urquijo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Xavier Gotsens
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
| | - Karine Fauria
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Jordi Sunyer
- Universitat Pompeu Fabra (UPF), Barcelona, Spain; ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Mark J Nieuwenhuijsen
- Universitat Pompeu Fabra (UPF), Barcelona, Spain; ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| |
Collapse
|
142
|
Uddin MS, Kabir MT, Jeandet P, Mathew B, Ashraf GM, Perveen A, Bin-Jumah MN, Mousa SA, Abdel-Daim MM. Novel Anti-Alzheimer's Therapeutic Molecules Targeting Amyloid Precursor Protein Processing. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7039138. [PMID: 32411333 PMCID: PMC7206886 DOI: 10.1155/2020/7039138] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 03/27/2020] [Accepted: 04/01/2020] [Indexed: 02/04/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia among older people, and the prevalence of this disease is estimated to rise quickly in the upcoming years. Unfortunately, almost all of the drug candidates tested for AD until now have failed to exhibit any efficacy. Henceforth, there is an increased necessity to avert and/or slow down the advancement of AD. It is known that one of the major pathological characteristics of AD is the presence of senile plaques (SPs) in the brain. These SPs are composed of aggregated amyloid beta (Aβ), derived from the amyloid precursor protein (APP). Pharmaceutical companies have conducted a number of studies in order to identify safe and effective anti-Aβ drugs to combat AD. It is known that α-, β-, and γ-secretases are the three proteases that are involved in APP processing. Furthermore, there is a growing interest in these proteases, as they have a contribution to the modulation and production of Aβ. It has been observed that small compounds can be used to target these important proteases. Indeed, these compounds must satisfy the common strict requirements of a drug candidate targeted for brain penetration and selectivity toward different proteases. In this article, we have focused on the auspicious molecules which are under development for targeting APP-processing enzymes. We have also presented several anti-AD molecules targeting Aβ accumulation and phosphorylation signaling in APP processing. This review highlights the structure-activity relationship and other physicochemical features of several pharmacological candidates in order to successfully develop new anti-AD drugs.
Collapse
Affiliation(s)
- Md. Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | | | - Philippe Jeandet
- Research Unit, Induced Resistance and Plant Bioprotection, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, University of Reims Champagne-Ardenne, PO Box 1039, 51687 Reims Cedex 2, France
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - May N. Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia
| | - Shaker A. Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, New York, NY 12144, USA
| | - Mohamed M. Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
143
|
Huang SY, Zhu JX, Shen XN, Xu W, Ma YH, Li HQ, Dong Q, Tan L, Yu JT. Prevalence of the Preclinical Stages of Alzheimer's Disease in Cognitively Intact Older Adults: The CABLE Study. J Alzheimers Dis 2020; 75:483-492. [PMID: 32310174 DOI: 10.3233/jad-200059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The National Institute on Aging and Alzheimer's Association proposed an ATN classification system which divided Alzheimer's disease biomarkers into three binary classes: amyloid deposition (A), tauopathy (T), and neurodegeneration or neuronal injury (N). OBJECTIVE To estimate the prevalence of each profile and to describe the demographic characteristics of each group in Chinese cognitively intact older adults. METHODS In this cross-sectional study, 561 cognitively intact participants from the Chinese Alzheimer's Biomarker and LifestylE (CABLE) study were classified into eight groups using cerebrospinal fluid amyloid-β 42/40 as A, phosphorylated tau as T, and total tau as N. Multinomial models were used to determine the estimated prevalence of the eight groups. RESULTS The number and proportion of 561 participants in each ATN profile were 254 A-T-N- (45.3%), 28 A-T+N- (5.0%), 21 A-T-N+ (3.7%), 71 A-T+N+ (12.7%), 78 A + T-N- (13.9%), 14 A + T+N- (2.5%), 21 A + T-N+ (3.7%), and 74 A + T+N+ (13.2%). Individuals in N+ groups tend to be older than N- groups. A+ groups included more female individuals. The prevalence of A-T-N- profile declined with age, while that of A + T+N+ increased continuously. CONCLUSION This is the first work to estimate the prevalence of each ATN profile and describe the demographic characteristics of ATN profiles based on a Chinese cohort. The clinical implications of our findings need to be scrutinized further in longitudinal studies of the ATN classification system.
Collapse
Affiliation(s)
- Shu-Yi Huang
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jun-Xia Zhu
- Department of Prevention and Health Protection, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xue-Ning Shen
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Xu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Hong-Qi Li
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
144
|
Janelidze S, Stomrud E, Smith R, Palmqvist S, Mattsson N, Airey DC, Proctor NK, Chai X, Shcherbinin S, Sims JR, Triana-Baltzer G, Theunis C, Slemmon R, Mercken M, Kolb H, Dage JL, Hansson O. Cerebrospinal fluid p-tau217 performs better than p-tau181 as a biomarker of Alzheimer's disease. Nat Commun 2020; 11:1683. [PMID: 32246036 PMCID: PMC7125218 DOI: 10.1038/s41467-020-15436-0] [Citation(s) in RCA: 277] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 03/06/2020] [Indexed: 01/01/2023] Open
Abstract
Cerebrospinal fluid (CSF) p-tau181 (tau phosphorylated at threonine 181) is an established biomarker of Alzheimer's disease (AD), reflecting abnormal tau metabolism in the brain. Here we investigate the performance of CSF p-tau217 as a biomarker of AD in comparison to p-tau181. In the Swedish BioFINDER cohort (n = 194), p-tau217 shows stronger correlations with the tau positron emission tomography (PET) tracer [18F]flortaucipir, and more accurately identifies individuals with abnormally increased [18F]flortaucipir retention. Furthermore, longitudinal increases in p-tau217 are higher compared to p-tau181 and better correlate with [18F]flortaucipir uptake. P-tau217 correlates better than p-tau181 with CSF and PET measures of neocortical amyloid-β burden and more accurately distinguishes AD dementia from non-AD neurodegenerative disorders. Higher correlations between p-tau217 and [18F]flortaucipir are corroborated in an independent EXPEDITION3 trial cohort (n = 32). The main results are validated using a different p-tau217 immunoassay. These findings suggest that p-tau217 might be more useful than p-tau181 in the diagnostic work up of AD.
Collapse
Affiliation(s)
- Shorena Janelidze
- Clinical Memory Research Unit, Lund University, Sölvegatan 18, Lund, Sweden.
| | - Erik Stomrud
- Clinical Memory Research Unit, Lund University, Sölvegatan 18, Lund, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Lund University, Sölvegatan 18, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Entrégatan 7, 222 42, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Lund University, Sölvegatan 18, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Entrégatan 7, 222 42, Lund, Sweden
| | - Niklas Mattsson
- Clinical Memory Research Unit, Lund University, Sölvegatan 18, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Entrégatan 7, 222 42, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Klinikgatan 32, 221 84, Lund, Sweden
| | | | | | - Xiyun Chai
- Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | | | - John R Sims
- Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Gallen Triana-Baltzer
- Neuroscience Biomarkers, Janssen Research & Development, 3210 Merryfield Row, San Diego, CA, CA 92121, USA
| | - Clara Theunis
- Janssen Pharmaceutical Companies of Johnson & Johnson, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Randy Slemmon
- Neuroscience Biomarkers, Janssen Research & Development, 3210 Merryfield Row, San Diego, CA, CA 92121, USA
| | - Marc Mercken
- Janssen Pharmaceutical Companies of Johnson & Johnson, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Hartmuth Kolb
- Neuroscience Biomarkers, Janssen Research & Development, 3210 Merryfield Row, San Diego, CA, CA 92121, USA.
| | | | - Oskar Hansson
- Clinical Memory Research Unit, Lund University, Sölvegatan 18, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Simrisbanvägen 14, 205 02, Malmö, Sweden.
| |
Collapse
|
145
|
Babulal GM, Stout SH, Williams MM, Rajasekar G, Harmon A, Vivoda J, Zuelsdorff M, Benzinger TLS, Morris JC, Ances B, Roe CM. Differences in Driving Outcomes Among Cognitively Normal African American and Caucasian Older Adults. J Racial Ethn Health Disparities 2020; 7:269-280. [PMID: 31686371 PMCID: PMC7067633 DOI: 10.1007/s40615-019-00655-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/11/2019] [Accepted: 10/14/2019] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To examine the effect of race in driving performance and behavior prospectively among cognitively normal older adults. METHODS Cognitively normal participants (Clinical Dementia Rating 0), ≥ 65 years of age (n = 177) were selected from prospective, longitudinal studies at the Knight Alzheimer Disease Research Center at Washington University. Self-reported driving behavior (Driving Habits Questionnaire) and driving performance (road test) were annually assessed. Daily driving behavior data were collected using the Driving Real World In-Vehicle Evaluation System (DRIVES). Baseline differences between African Americans and Caucasians were tested using t tests and general linear models. Amyloid imaging and cerebrospinal fluid Alzheimer disease (AD) biomarkers were compared across groups. Linear mixed models examined change in daily driving behavior over time. Survival analyses tested time to a marginal or fail rating on the road test. RESULTS There were no differences between African Americans (n = 34) and Caucasians (n = 143) in age, sex, education, or vascular risk factors. Baseline self-reported driving behavior and road test performance were largely similar for both races. Longitudinal analyses using the DRIVES data aggregated monthly showed that African Americans had a greater reduction in number of trips made per month, miles driven per month, and trips with aggressive behavior compared to Caucasians. These effects remained after controlling for AD biomarkers, age, education, and sex. CONCLUSIONS In this sample of cognitively normal older adults, African Americans had a greater reduction of daily driving behavior compared to Caucasians. Observed racial differences may reflect differences in environmental/social factors, changes in cognition, and/or physical functioning.
Collapse
Affiliation(s)
- Ganesh M Babulal
- Charles F. and Joanne Knight Alzheimer's Disease Research Center, 660 S. Euclid Ave., Campus Box, St. Louis, MO, 8111, USA.
- Department of Neurology, Washington University, St. Louis, MO, USA.
| | - Sarah H Stout
- Charles F. and Joanne Knight Alzheimer's Disease Research Center, 660 S. Euclid Ave., Campus Box, St. Louis, MO, 8111, USA
- Department of Neurology, Washington University, St. Louis, MO, USA
| | | | - Ganesh Rajasekar
- Charles F. and Joanne Knight Alzheimer's Disease Research Center, 660 S. Euclid Ave., Campus Box, St. Louis, MO, 8111, USA
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - Annie Harmon
- Department of Medicine, Washington University, St. Louis, MO, USA
| | | | | | - Tammie L S Benzinger
- Charles F. and Joanne Knight Alzheimer's Disease Research Center, 660 S. Euclid Ave., Campus Box, St. Louis, MO, 8111, USA
- Department of Radiology, Washington University, St. Louis, MO, USA
- Department of Neurosurgery, Washington University, St. Louis, MO, USA
| | - John C Morris
- Charles F. and Joanne Knight Alzheimer's Disease Research Center, 660 S. Euclid Ave., Campus Box, St. Louis, MO, 8111, USA
- Department of Neurology, Washington University, St. Louis, MO, USA
- Department of Radiology, Washington University, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University, St. Louis, MO, USA
- Department of Physical Therapy, Washington University, St. Louis, MO, USA
- Department of Occupational Therapy, Washington University, St. Louis, MO, USA
| | - Beau Ances
- Charles F. and Joanne Knight Alzheimer's Disease Research Center, 660 S. Euclid Ave., Campus Box, St. Louis, MO, 8111, USA
- Department of Neurology, Washington University, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University, St. Louis, MO, USA
| | - Catherine M Roe
- Charles F. and Joanne Knight Alzheimer's Disease Research Center, 660 S. Euclid Ave., Campus Box, St. Louis, MO, 8111, USA
- Department of Neurology, Washington University, St. Louis, MO, USA
| |
Collapse
|
146
|
Profile of Pathogenic Proteins and MicroRNAs in Plasma-derived Extracellular Vesicles in Alzheimer’s Disease: A Pilot Study. Neuroscience 2020; 432:240-246. [DOI: 10.1016/j.neuroscience.2020.02.044] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 02/23/2020] [Accepted: 02/24/2020] [Indexed: 12/19/2022]
|
147
|
Singh AK, Verma S. Use of ocular biomarkers as a potential tool for early diagnosis of Alzheimer's disease. Indian J Ophthalmol 2020; 68:555-561. [PMID: 32174567 PMCID: PMC7210832 DOI: 10.4103/ijo.ijo_999_19] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/10/2019] [Accepted: 10/26/2019] [Indexed: 02/05/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide which unfortunately has no known effective cure to date. Despite many clinical trials indicating the effectiveness of preclinical treatment, a sensitive tool for screening of AD is yet to be developed. Due to multiple similarities between ocular and the brain tissue, the eye is being explored by researchers for this purpose, with utmost attention focused on the retinal tissue. Besides visual functional impairment, neuronal degeneration and apoptosis, retinal nerve fiber degeneration, increase in the cup-to-disc ratio, and retinal vascular thinning and tortuosity are the changes observed in the retinal tissue which are related to AD. Studies have shown that targeting these changes in the retina is an effective way of reducing the degeneration of retinal neuronal tissue. Similar mechanisms of neurodegeneration have been demonstrated in the brain and the eyes of AD patients. Multiple studies are underway to investigate the potential of diagnosing AD and detection of amyloid-β (Aβ) levels in the retinal tissue. Since the tissues in the anterior segment of the eye are more accessible for in vivo imaging and examination, they have more potential as screening biomarkers. This article provides a concise review of available literature on the ocular biomarkers in anterior and posterior segments of the eye including the cornea, aqueous humour (AH), crystalline lens, and retina in AD. This review will also highlight the newer technological tools available for the detection of potential biomarkers in the eye for early diagnosis of AD.
Collapse
Affiliation(s)
- Ajay K Singh
- Consultant and Anterior Segment Surgeon, Department of Ophthalmology, Asian Institute of Medical Sciences, Faridabad, Haryana, India
| | - Shilpa Verma
- WNS Global Services Pvt. Ltd., Gurugram, Haryana, India
| |
Collapse
|
148
|
Examining the Complicated Relationship Between Depressive Symptoms and Cognitive Impairment in Preclinical Alzheimer Disease. Alzheimer Dis Assoc Disord 2020; 33:15-20. [PMID: 30489279 DOI: 10.1097/wad.0000000000000284] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION The relationships between Alzheimer disease (AD), cognitive performance, and depression are poorly understood. It is unclear whether depressive features are a prodrome of AD. In addition, some studies of aging exclude depressed individuals, which may inappropriately limit generalizability. The aim of the present study was to determine whether depressive symptoms affect cognitive function in the context of preclinical AD. METHODS Cross-sectional multivariate analysis of participants in a longitudinal study of aging (n=356) that evaluates the influence of depressive symptoms on cognitive function in cognitively normal adults. RESULTS There is no relationship between the presence of depressive symptoms and cognitive function in those with either no evidence of preclinical AD or biomarker evidence of early-stage preclinical AD. However, in later stages of preclinical AD, the presence of depressive symptoms demonstrated interactive effects, including in episodic memory (0.96; 95% confidence interval, 0.31-1.62) and global cognitive function (0.46; 95% confidence interval, 0.028-0.89). CONCLUSIONS The presence of depressive symptoms may be a late prodrome of AD. In addition, studies investigating cognitive function in older adults may not need to exclude participants with depressive symptomology, but may still consider depressive symptoms as a potential confounder in the context of more extensive neuronal injury.
Collapse
|
149
|
Barthélemy NR, Bateman RJ, Hirtz C, Marin P, Becher F, Sato C, Gabelle A, Lehmann S. Cerebrospinal fluid phospho-tau T217 outperforms T181 as a biomarker for the differential diagnosis of Alzheimer's disease and PET amyloid-positive patient identification. Alzheimers Res Ther 2020; 12:26. [PMID: 32183883 PMCID: PMC7079453 DOI: 10.1186/s13195-020-00596-4] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 03/09/2020] [Indexed: 11/14/2023]
Abstract
BACKGROUND Cerebrospinal fluid biomarker profiles characterized by decreased amyloid-beta peptide levels and increased total and phosphorylated tau levels at threonine 181 (pT181) are currently used to discriminate between Alzheimer's disease and other neurodegenerative diseases. However, these changes are not entirely specific to Alzheimer's disease, and it is noteworthy that other phosphorylated isoforms of tau, possibly more specific for the disease process, have been described in the brain parenchyma of patients. The precise detection of these isoforms in biological fluids remains however a challenge. METHODS In the present study, we used the latest quantitative mass spectrometry approach, which achieves a sensitive detection in cerebrospinal fluid biomarker of two phosphorylated tau isoforms, pT181 and pT217, and first analyzed a cohort of probable Alzheimer's disease patients and patients with other neurological disorders, including tauopathies, and a set of cognitively normal controls. We then checked the validity of our results on a second cohort comprising cognitively normal individuals and patients with mild cognitive impairments and AD stratified in terms of their amyloid status based on PiB-PET imaging methods. RESULTS In the first cohort, pT217 but not pT181 differentiated between Alzheimer's disease patients and those with other neurodegenerative diseases and control subjects much more specificity and sensitivity than pT181. T217 phosphorylation was increased by 6.0-fold in patients with Alzheimer's disease whereas T181 phosphorylation was only increased by 1.3-fold, when compared with control subjects. These results were confirmed in the case of a second cohort, in which the pT217 cerebrospinal fluid levels marked out amyloid-positive patients with a sensitivity and a specificity of more than 90% (AUC 0.961; CI 0.874 to 0.995). The pT217 concentrations were also highly correlated with the PiB-PET values (correlation coefficient 0.72; P < 0.001). CONCLUSIONS Increased cerebrospinal fluid pT217 levels, more than those of pT181, are highly specific biomarkers for detecting both the preclinical and advanced forms of Alzheimer's disease. This finding should greatly improve the diagnosis of Alzheimer's disease, along with the correlations found to exist between pT217 levels and PiB-PET data. It also suggests that pT217 is a promising potential target for therapeutic applications and that a link exists between amyloid and tau pathology.
Collapse
Affiliation(s)
- Nicolas R Barthélemy
- Laboratoire de Biochimie Protéomique Clinique, Plateforme de Protéomique Clinique, CHU de Montpellier, INSERM, Université de Montpellier, Montpellier, France.
- Department of Neurology, Washington University School of Medicine, Saint-Louis, MO, USA.
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, Saint-Louis, MO, USA
| | - Christophe Hirtz
- Laboratoire de Biochimie Protéomique Clinique, Plateforme de Protéomique Clinique, CHU de Montpellier, INSERM, Université de Montpellier, Montpellier, France
| | - Philippe Marin
- Institut de Génomique Fonctionnelle, CNRS UMR5203, INSERM U1191, Université de Montpellier, Montpellier, France
| | - François Becher
- Service de Pharmacologie et d'Immunoanalyse (SPI), Laboratoire d'Etude du Métabolisme des Médicaments (LEMM), CEA, INRA, Université Paris Saclay, F-91191, Gif-sur-Yvette cedex, France
| | - Chihiro Sato
- Department of Neurology, Washington University School of Medicine, Saint-Louis, MO, USA
| | - Audrey Gabelle
- Memory Resources and Research Center of Montpellier, Department of Neurology, Gui de Chauliac Hospital, Montpellier, France.
| | - Sylvain Lehmann
- Laboratoire de Biochimie Protéomique Clinique, Plateforme de Protéomique Clinique, CHU de Montpellier, INSERM, Université de Montpellier, Montpellier, France.
| |
Collapse
|
150
|
Quevenco FC, van Bergen JM, Treyer V, Studer ST, Kagerer SM, Meyer R, Gietl AF, Kaufmann PA, Nitsch RM, Hock C, Unschuld PG. Functional Brain Network Connectivity Patterns Associated With Normal Cognition at Old-Age, Local β-amyloid, Tau, and APOE4. Front Aging Neurosci 2020; 12:46. [PMID: 32210782 PMCID: PMC7075450 DOI: 10.3389/fnagi.2020.00046] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 02/10/2020] [Indexed: 12/30/2022] Open
Abstract
Background: Integrity of functional brain networks is closely associated with maintained cognitive performance at old age. Consistently, both carrier status of Apolipoprotein E ε4 allele (APOE4), and age-related aggregation of Alzheimer’s disease (AD) pathology result in altered brain network connectivity. The posterior cingulate and precuneus (PCP) is a node of particular interest due to its role in crucial memory processes. Moreover, the PCP is subject to the early aggregation of AD pathology. The current study aimed at characterizing brain network properties associated with unimpaired cognition in old aged adults. To determine the effects of age-related brain change and genetic risk for AD, pathological proteins β-amyloid and tau were measured by Positron-emission tomography (PET), PCP connectivity as a proxy of cognitive network integrity, and genetic risk by APOE4 carrier status. Methods: Fifty-seven cognitively unimpaired old-aged adults (MMSE = 29.20 ± 1.11; 73 ± 8.32 years) were administered 11C Pittsburgh Compound B and 18F Flutemetamol PET for assessing β-amyloid, and 18F AV-1451 PET for tau. Individual functional connectivity seed maps of the PCP were obtained by resting-state multiband BOLD functional MRI at 3-Tesla for increased temporal resolution. Voxelwise correlations between functional connectivity, β-amyloid- and tau-PET were explored by Biological Parametric Mapping (BPM). Results: Local β-amyloid was associated with increased connectivity in frontal and parietal regions of the brain. Tau was linked to increased connectivity in more spatially distributed clusters in frontal, parietal, occipital, temporal, and cerebellar regions. A positive interaction was observable for APOE4 carrier status and functional connectivity with brain regions characterized by increased local β-amyloid and tau tracer retention. Conclusions: Our data suggest an association between spatially differing connectivity systems and local β-amyloid, and tau aggregates in cognitively normal, old-aged adults, which is moderated by APOE4. Additional longitudinal studies may determine protective connectivity patterns associated with healthy aging trajectories of AD-pathology aggregation.
Collapse
Affiliation(s)
- Frances C Quevenco
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Jiri M van Bergen
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Valerie Treyer
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland.,Department of Nuclear Medicine, University of Zurich, Zurich, Switzerland
| | - Sandro T Studer
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Sonja M Kagerer
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland.,Neurimmune, Schlieren, Switzerland
| | - Rafael Meyer
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Anton F Gietl
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Philipp A Kaufmann
- Department of Nuclear Medicine, University of Zurich, Zurich, Switzerland
| | - Roger M Nitsch
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland.,Neurimmune, Schlieren, Switzerland
| | - Christoph Hock
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland.,Neurimmune, Schlieren, Switzerland
| | - Paul G Unschuld
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland.,Department of Psychogeriatric Medicine, Psychiatric University Hospital Zurich (PUK), Zurich, Switzerland.,Zurich Neuroscience Center (ZNZ), Zurich, Switzerland
| |
Collapse
|