51
|
Kang JH, Korecka M, Lee EB, Cousins KAQ, Tropea TF, Chen-Plotkin AA, Irwin DJ, Wolk D, Brylska M, Wan Y, Shaw LM. Alzheimer Disease Biomarkers: Moving from CSF to Plasma for Reliable Detection of Amyloid and tau Pathology. Clin Chem 2023; 69:1247-1259. [PMID: 37725909 PMCID: PMC10895336 DOI: 10.1093/clinchem/hvad139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 07/07/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Development of validated biomarkers to detect early Alzheimer disease (AD) neuropathology is needed for therapeutic AD trials. Abnormal concentrations of "core" AD biomarkers, cerebrospinal fluid (CSF) amyloid beta1-42, total tau, and phosphorylated tau correlate well with neuroimaging biomarkers and autopsy findings. Nevertheless, given the limitations of established CSF and neuroimaging biomarkers, accelerated development of blood-based AD biomarkers is underway. CONTENT Here we describe the clinical significance of CSF and plasma AD biomarkers to detect disease pathology throughout the Alzheimer continuum and correlate with imaging biomarkers. Use of the AT(N) classification by CSF and imaging biomarkers provides a more objective biologically based diagnosis of AD than clinical diagnosis alone. Significant progress in measuring CSF AD biomarkers using extensively validated highly automated assay systems has facilitated their transition from research use only to approved in vitro diagnostics tests for clinical use. We summarize development of plasma AD biomarkers as screening tools for enrollment and monitoring participants in therapeutic trials and ultimately in clinical care. Finally, we discuss the challenges for AD biomarkers use in clinical trials and precision medicine, emphasizing the possible ethnocultural differences in the levels of AD biomarkers. SUMMARY CSF AD biomarker measurements using fully automated analytical platforms is possible. Building on this experience, validated blood-based biomarker tests are being implemented on highly automated immunoassay and mass spectrometry platforms. The progress made developing analytically and clinically validated plasma AD biomarkers within the AT(N) classification scheme can accelerate use of AD biomarkers in therapeutic trials and routine clinical practice.
Collapse
Affiliation(s)
- Ju Hee Kang
- Department of Pharmacology and Clinical Pharmacology, Research Center for Controlling Intercellular Communication, Inha University, Incheon, South Korea
| | - Magdalena Korecka
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Katheryn A Q Cousins
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Thomas F Tropea
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Alice A Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - David Wolk
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Magdalena Brylska
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yang Wan
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
52
|
Lew CO, Zhou L, Mazurowski MA, Doraiswamy PM, Petrella JR. MRI-based Deep Learning Assessment of Amyloid, Tau, and Neurodegeneration Biomarker Status across the Alzheimer Disease Spectrum. Radiology 2023; 309:e222441. [PMID: 37815445 PMCID: PMC10623183 DOI: 10.1148/radiol.222441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 09/03/2023] [Accepted: 09/11/2023] [Indexed: 10/11/2023]
Abstract
Background PET can be used for amyloid-tau-neurodegeneration (ATN) classification in Alzheimer disease, but incurs considerable cost and exposure to ionizing radiation. MRI currently has limited use in characterizing ATN status. Deep learning techniques can detect complex patterns in MRI data and have potential for noninvasive characterization of ATN status. Purpose To use deep learning to predict PET-determined ATN biomarker status using MRI and readily available diagnostic data. Materials and Methods MRI and PET data were retrospectively collected from the Alzheimer's Disease Imaging Initiative. PET scans were paired with MRI scans acquired within 30 days, from August 2005 to September 2020. Pairs were randomly split into subsets as follows: 70% for training, 10% for validation, and 20% for final testing. A bimodal Gaussian mixture model was used to threshold PET scans into positive and negative labels. MRI data were fed into a convolutional neural network to generate imaging features. These features were combined in a logistic regression model with patient demographics, APOE gene status, cognitive scores, hippocampal volumes, and clinical diagnoses to classify each ATN biomarker component as positive or negative. Area under the receiver operating characteristic curve (AUC) analysis was used for model evaluation. Feature importance was derived from model coefficients and gradients. Results There were 2099 amyloid (mean patient age, 75 years ± 10 [SD]; 1110 male), 557 tau (mean patient age, 75 years ± 7; 280 male), and 2768 FDG PET (mean patient age, 75 years ± 7; 1645 male) and MRI pairs. Model AUCs for the test set were as follows: amyloid, 0.79 (95% CI: 0.74, 0.83); tau, 0.73 (95% CI: 0.58, 0.86); and neurodegeneration, 0.86 (95% CI: 0.83, 0.89). Within the networks, high gradients were present in key temporal, parietal, frontal, and occipital cortical regions. Model coefficients for cognitive scores, hippocampal volumes, and APOE status were highest. Conclusion A deep learning algorithm predicted each component of PET-determined ATN status with acceptable to excellent efficacy using MRI and other available diagnostic data. © RSNA, 2023 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Christopher O. Lew
- From the Department of Radiology, Division of Neuroradiology,
Alzheimer Disease Imaging Research Laboratory (C.O.L., J.R.P.), and
Neurocognitive Disorders Program, Departments of Psychiatry and Medicine
(P.M.D.), Duke University Medical Center, DUMC-Box 3808, Durham, NC 27710-3808;
and Duke Institute for Brain Sciences (P.M.D.) and Department of Electrical and
Computer Engineering, Department of Computer Science, Department of
Biostatistics and Bioinformatics (L.Z., M.A.M.), Duke University, Durham,
NC
| | - Longfei Zhou
- From the Department of Radiology, Division of Neuroradiology,
Alzheimer Disease Imaging Research Laboratory (C.O.L., J.R.P.), and
Neurocognitive Disorders Program, Departments of Psychiatry and Medicine
(P.M.D.), Duke University Medical Center, DUMC-Box 3808, Durham, NC 27710-3808;
and Duke Institute for Brain Sciences (P.M.D.) and Department of Electrical and
Computer Engineering, Department of Computer Science, Department of
Biostatistics and Bioinformatics (L.Z., M.A.M.), Duke University, Durham,
NC
| | - Maciej A. Mazurowski
- From the Department of Radiology, Division of Neuroradiology,
Alzheimer Disease Imaging Research Laboratory (C.O.L., J.R.P.), and
Neurocognitive Disorders Program, Departments of Psychiatry and Medicine
(P.M.D.), Duke University Medical Center, DUMC-Box 3808, Durham, NC 27710-3808;
and Duke Institute for Brain Sciences (P.M.D.) and Department of Electrical and
Computer Engineering, Department of Computer Science, Department of
Biostatistics and Bioinformatics (L.Z., M.A.M.), Duke University, Durham,
NC
| | - P. Murali Doraiswamy
- From the Department of Radiology, Division of Neuroradiology,
Alzheimer Disease Imaging Research Laboratory (C.O.L., J.R.P.), and
Neurocognitive Disorders Program, Departments of Psychiatry and Medicine
(P.M.D.), Duke University Medical Center, DUMC-Box 3808, Durham, NC 27710-3808;
and Duke Institute for Brain Sciences (P.M.D.) and Department of Electrical and
Computer Engineering, Department of Computer Science, Department of
Biostatistics and Bioinformatics (L.Z., M.A.M.), Duke University, Durham,
NC
| | - Jeffrey R. Petrella
- From the Department of Radiology, Division of Neuroradiology,
Alzheimer Disease Imaging Research Laboratory (C.O.L., J.R.P.), and
Neurocognitive Disorders Program, Departments of Psychiatry and Medicine
(P.M.D.), Duke University Medical Center, DUMC-Box 3808, Durham, NC 27710-3808;
and Duke Institute for Brain Sciences (P.M.D.) and Department of Electrical and
Computer Engineering, Department of Computer Science, Department of
Biostatistics and Bioinformatics (L.Z., M.A.M.), Duke University, Durham,
NC
| | | |
Collapse
|
53
|
Wang J, Chen M, Masters CL, Wang YJ. Translating blood biomarkers into clinical practice for Alzheimer's disease: Challenges and perspectives. Alzheimers Dement 2023; 19:4226-4236. [PMID: 37218404 DOI: 10.1002/alz.13116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 05/24/2023]
Abstract
Early and accurate diagnosis of Alzheimer's disease (AD) in clinical practice is urgent with advances in AD treatment. Blood biomarker assays are preferential diagnostic tools for widespread clinical use with the advantages of being less invasive, cost effective, and easily accessible, and they have shown good performance in research cohorts. However, in community-based populations with maximum heterogeneity, great challenges are still faced in diagnosing AD based on blood biomarkers in terms of accuracy and robustness. Here, we analyze these challenges, including the confounding impact of systemic and biological factors, small changes in blood biomarkers, and difficulty in detecting early changes. Furthermore, we provide perspectives on several potential strategies to overcome these challenges for blood biomarkers to bridge the gap from research to clinical practice.
Collapse
Affiliation(s)
- Jun Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Ming Chen
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Yan-Jiang Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
- State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
54
|
Mourtzi N, Charisis S, Tsapanou A, Ntanasi E, Hatzimanolis A, Ramirez A, Heilmann-Heimbach S, Grenier-Boley B, Lambert JC, Yannakoulia M, Kosmidis M, Dardiotis E, Hadjigeorgiou G, Sakka P, Georgakis M, Yaakov S, Scarmeas N. Genetic propensity for cerebral amyloidosis and risk of mild cognitive impairment and Alzheimer's disease within a cognitive reserve framework. Alzheimers Dement 2023; 19:3794-3805. [PMID: 36895094 DOI: 10.1002/alz.12980] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/29/2022] [Accepted: 01/16/2023] [Indexed: 03/11/2023]
Abstract
INTRODUCTION We constructed a polygenic risk score (PRS) for β-amyloid (PRSAβ42) to proxy AD pathology and investigated its association with incident Alzheimer's disease (AD)/amnestic mild cognitive impairment (aMCI) and the influence of cognitive reserve (CR), proxied by educational years, on the relationship between PRSAβ42 and AD/aMCI risk. METHODS A total of 618 cognitive-normal participants were followed-up for 2.92 years. The association of PRSAβ42 and CR with AD/aMCI incidence was examined with COX models. Then we examined the additive interaction between PRSAβ42 and CR and the CR effect across participants with different PRSAβ42 levels. RESULTS Higher PRSAβ42 and CR were associated with a 33.9% higher risk and 8.3% less risk for AD/aMCI, respectively. An additive interaction between PRSAβ42 and CR was observed. High CR was associated with 62.6% less risk of AD/aMCI incidence only in the high-PRSAβ42 group. DISCUSSION A super-additive effect of PRSAβ42 and CR on AD/aMCI risk was observed. CR influence was evident in participants with high PRSAβ42.
Collapse
Affiliation(s)
- Niki Mourtzi
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Sokratis Charisis
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
- Department of Neurology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Angeliki Tsapanou
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
- Department of Neurology, The Gertrude H. Sergievsky Center, Taub Institute for Research in Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, USA
| | - Eva Ntanasi
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | - Alexandros Hatzimanolis
- Department of Psychiatry, National and Kapodistrian University of Athens Medical School, Eginition Hospital, Athens, Greece
| | - Alfredo Ramirez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE Bonn), Bonn, Germany
- Department of Psychiatry, Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, Texas, USA
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Stefanie Heilmann-Heimbach
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Benjamin Grenier-Boley
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liés au vieillissement, Lille, France
| | - Jean-Charles Lambert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liés au vieillissement, Lille, France
| | - Mary Yannakoulia
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | - Mary Kosmidis
- Lab of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Efthimios Dardiotis
- Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | | | - Paraskevi Sakka
- Athens Association of Alzheimer's Disease and Related Disorders, Marousi, Greece
| | - Marios Georgakis
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and the Massachusetts Institute of Technology, Boston, Massachusetts, USA
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Stern Yaakov
- Department of Neurology, The Gertrude H. Sergievsky Center, Taub Institute for Research in Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, USA
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
- Department of Neurology, The Gertrude H. Sergievsky Center, Taub Institute for Research in Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, USA
| |
Collapse
|
55
|
Nisenbaum L, Martone R, Chen T, Rajagovindan R, Dent G, Beaver J, Rubel C, Racine A, He P, Harrison K, Dean R, Vandijck M, Haeberlein SB. CSF biomarker concordance with amyloid PET in Phase 3 studies of aducanumab. Alzheimers Dement 2023; 19:3379-3388. [PMID: 36795603 DOI: 10.1002/alz.12919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 02/17/2023]
Abstract
INTRODUCTION We assessed the use of cerebrospinal fluid (CSF) biomarkers as an alternative to positron emission tomography (PET) for brain amyloid beta (Aβ) pathology confirmation in the EMERGE and ENGAGE clinical trials. METHODS EMERGE and ENGAGE were randomized, placebo-controlled, Phase 3 trials of aducanumab in participants with early Alzheimer's disease. Concordance between CSF biomarkers (Aβ42, Aβ40, phosphorylated tau 181, and total tau) and amyloid PET status (visual read) at screening was examined. RESULTS Robust concordance between CSF biomarkers and amyloid PET visual status was observed (for Aβ42/Aβ40, AUC: 0.90; 95% CI: 0.83-0.97; p < 0.0001), confirming CSF biomarkers as a reliable alternative to amyloid PET in these studies. Compared with single CSF biomarkers, CSF biomarker ratios showed better agreement with amyloid PET visual reads, demonstrating high diagnostic accuracy. DISCUSSION These analyses add to the growing body of evidence supporting CSF biomarkers as reliable alternatives to amyloid PET imaging for brain Aβ pathology confirmation. HIGHLIGHTS CSF biomarkers and amyloid PET concordance were assessed in Ph3 aducanumab trials. Robust concordance between CSF biomarkers and amyloid PET was observed. CSF biomarker ratios increased diagnostic accuracy over single CSF biomarkers. CSF Aβ42/Aβ40 demonstrated high concordance with amyloid PET. Results support CSF biomarker testing as a reliable alternative to amyloid PET.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ping He
- Biogen, Cambridge, Massachusetts, USA
| | | | - Robert Dean
- Robert A. Dean Consulting, LLC, Indianapolis, Indiana, USA
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | |
Collapse
|
56
|
Wang Y, Wu S, Li Q, Sun H, Wang H. Pharmacological Inhibition of Ferroptosis as a Therapeutic Target for Neurodegenerative Diseases and Strokes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300325. [PMID: 37341302 PMCID: PMC10460905 DOI: 10.1002/advs.202300325] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 05/23/2023] [Indexed: 06/22/2023]
Abstract
Emerging evidence suggests that ferroptosis, a unique regulated cell death modality that is morphologically and mechanistically different from other forms of cell death, plays a vital role in the pathophysiological process of neurodegenerative diseases, and strokes. Accumulating evidence supports ferroptosis as a critical factor of neurodegenerative diseases and strokes, and pharmacological inhibition of ferroptosis as a therapeutic target for these diseases. In this review article, the core mechanisms of ferroptosis are overviewed and the roles of ferroptosis in neurodegenerative diseases and strokes are described. Finally, the emerging findings in treating neurodegenerative diseases and strokes through pharmacological inhibition of ferroptosis are described. This review demonstrates that pharmacological inhibition of ferroptosis by bioactive small-molecule compounds (ferroptosis inhibitors) could be effective for treatments of these diseases, and highlights a potential promising therapeutic avenue that could be used to prevent neurodegenerative diseases and strokes. This review article will shed light on developing novel therapeutic regimens by pharmacological inhibition of ferroptosis to slow down the progression of these diseases in the future.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care MedicineAerospace Center HospitalPeking University Aerospace School of Clinical MedicineBeijing100049P. R. China
| | - Shuang Wu
- Department of NeurologyZhongnan Hospital of Wuhan UniversityWuhan430000P. R. China
| | - Qiang Li
- Department of NeurologyThe Affiliated Hospital of Chifeng UniversityChifeng024005P. R. China
| | - Huiyan Sun
- Chifeng University Health Science CenterChifeng024000P. R. China
| | - Hongquan Wang
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060P. R. China
| |
Collapse
|
57
|
Buss SS, Fried PJ, Macone J, Zeng V, Zingg E, Santarnecchi E, Pascual-Leone A, Bartrés-Faz D. Greater cognitive reserve is related to lower cortical excitability in healthy cognitive aging, but not in early clinical Alzheimer's disease. Front Hum Neurosci 2023; 17:1193407. [PMID: 37576473 PMCID: PMC10413110 DOI: 10.3389/fnhum.2023.1193407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/05/2023] [Indexed: 08/15/2023] Open
Abstract
Objective To investigate the relationship between cortico-motor excitability and cognitive reserve (CR) in cognitively unimpaired older adults (CU) and in older adults with mild cognitive impairment or mild dementia due to Alzheimer's disease (AD). Methods Data were collected and analyzed from 15 CU and 24 amyloid-positive AD participants aged 50-90 years. A cognitive reserve questionnaire score (CRQ) assessed education, occupation, leisure activities, physical activities, and social engagement. Cortical excitability was quantified as the average amplitude of motor evoked potentials (MEP amplitude) elicited with single-pulse transcranial magnetic stimulation delivered to primary motor cortex. A linear model compared MEP amplitudes between groups. A linear model tested for an effect of CRQ on MEP amplitude across all participants. Finally, separate linear models tested for an effect of CRQ on MEP amplitude within each group. Exploratory analyses tested for effect modification of demographics, cognitive scores, atrophy measures, and CSF measures within each group using nested regression analysis. Results There was no between-group difference in MEP amplitude after accounting for covariates. The primary model showed a significant interaction term of group*CRQ (R2adj = 0.18, p = 0.013), but no main effect of CRQ. Within the CU group, higher CRQ was significantly associated with lower MEP amplitude (R2adj = 0.45, p = 0.004). There was no association in the AD group. Conclusion Lower cortico-motor excitability is related to greater CRQ in CU, but not in AD. Lower MEP amplitudes may reflect greater neural efficiency in cognitively unimpaired older adults. The lack of association seen in AD participants may reflect disruption of the protective effects of CR. Future work is needed to better understand the neurophysiologic mechanisms leading to the protective effects of CR in older adults with and without neurodegenerative disorders.
Collapse
Affiliation(s)
- Stephanie S. Buss
- Division of Cognitive Neurology, Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Boston, MA, United States
- Department of Neurology, Harvard Medical School, Boston, MA, United States
| | - Peter J. Fried
- Division of Cognitive Neurology, Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Boston, MA, United States
- Department of Neurology, Harvard Medical School, Boston, MA, United States
| | - Joanna Macone
- Division of Cognitive Neurology, Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Boston, MA, United States
- Department of Neurology, Harvard Medical School, Boston, MA, United States
| | - Victor Zeng
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Emma Zingg
- Division of Cognitive Neurology, Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Boston, MA, United States
- Program of All-inclusive Care for the Elderly (PACE), Cambridge Health Alliance, Cambridge, MA, United States
| | - Emiliano Santarnecchi
- Division of Cognitive Neurology, Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Boston, MA, United States
- Department of Neurology, Harvard Medical School, Boston, MA, United States
- Precision Neuromodulation Program, Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Alvaro Pascual-Leone
- Department of Neurology, Harvard Medical School, Boston, MA, United States
- Deanna and Sidney Wolk Center for Memory Health, Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, United States
| | - David Bartrés-Faz
- Division of Cognitive Neurology, Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Boston, MA, United States
- Department of Medicine, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
58
|
Ciou SH, Hsieh AH, Lin YX, Sei JL, Govindasamy M, Kuo CF, Huang CH. Sensitive label-free detection of the biomarker phosphorylated tau-217 protein in Alzheimer's disease using a graphene-based solution-gated field effect transistor. Biosens Bioelectron 2023; 228:115174. [PMID: 36933321 DOI: 10.1016/j.bios.2023.115174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/18/2023] [Accepted: 02/18/2023] [Indexed: 03/13/2023]
Abstract
Alzheimer's disease (AD) is generally diagnosed using advanced imaging, but recent research suggests early screening using biomarkers in peripheral blood is feasible; among them, plasma tau proteins phosphorylated at threonine 231, threonine 181, and threonine 217 (p-tau217) are potential targets. A recent study indicates that the p-tau217 protein is the most efficacious biomarker. However, a clinical study found a pg/ml threshold for AD screening beyond standard detection methods. A biosensor with high sensitivity and specificity p-tau217 detection has not yet been reported. In this study, we developed a label-free solution-gated field effect transistor (SGFET)-based biosensor featuring a graphene oxide/graphene (GO/G) layered composite. The top layer of bilayer graphene grown using chemical vapor deposition was functionalized with oxidative groups serving as active sites for forming covalent bonds with the biorecognition element (antibodies); the bottom G could act as a transducer to respond to the attachment of the target analytes onto the top GO conjugated with the biorecognition element via π-π interactions between the GO and G layers. With this unique atomically layered G composite, we obtained a good linear electrical response in the Dirac point shift to p-tau217 protein concentrations in the range of 10 fg/ml to 100 pg/ml. The biosensor exhibited a high sensitivity of 18.6 mV/decade with a high linearity of 0.991 in phosphate-buffered saline (PBS); in human serum albumin, it showed approximately 90% of the sensitivity (16.7 mV/decade) in PBS, demonstrating high specificity. High stability of the biosensor was also displayed in this study.
Collapse
Affiliation(s)
- Sian-Hong Ciou
- Department of Materials Engineering, Ming Chi University of Technology, New Taipei City, 243303, Taiwan
| | - Ao-Ho Hsieh
- Novascope Diagnostics Inc., Taipei City, 10546, Taiwan
| | - Yu-Xiu Lin
- Department of Materials Engineering, Ming Chi University of Technology, New Taipei City, 243303, Taiwan
| | - Jhao-Liang Sei
- Department of Materials Engineering, Ming Chi University of Technology, New Taipei City, 243303, Taiwan
| | - Mani Govindasamy
- Department of Materials Engineering, Ming Chi University of Technology, New Taipei City, 243303, Taiwan
| | - Chang-Fu Kuo
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Taoyuan, 33305, Taiwan.
| | - Chi-Hsien Huang
- Department of Materials Engineering, Ming Chi University of Technology, New Taipei City, 243303, Taiwan; Novascope Diagnostics Inc., Taipei City, 10546, Taiwan.
| |
Collapse
|
59
|
Wu L, Xian X, Tan Z, Dong F, Xu G, Zhang M, Zhang F. The Role of Iron Metabolism, Lipid Metabolism, and Redox Homeostasis in Alzheimer's Disease: from the Perspective of Ferroptosis. Mol Neurobiol 2023; 60:2832-2850. [PMID: 36735178 DOI: 10.1007/s12035-023-03245-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/26/2023] [Indexed: 02/04/2023]
Abstract
In the development of Alzheimer's disease (AD), cell death is common. Novel cell death form-ferroptosis is discovered in recent years. Ferroptosis is an iron-regulated programmed cell death mechanism and has been identified in AD clinical samples. Typical characteristics of ferroptosis involve the specific changes in cell morphology, iron-dependent aggregation of reactive oxygen species (ROS) and lipid peroxides, loss of glutathione (GSH), inactivation of glutathione peroxidase 4 (GPX4), and a unique group of regulatory genes. Increasing evidence demonstrates that ferroptosis may be associated with neurological dysfunction in AD. However, the underlying mechanisms have not been fully elucidated. This article reviews the potential role of ferroptosis in AD, the involvement of ferroptosis in the pathological progression of AD through the mechanisms of iron metabolism, lipid metabolism, and redox homeostasis, as well as a range of potential therapies targeting ferroptosis for AD. Intervention strategies based on ferroptosis are promising for Alzheimer's disease treatment at present, but further researches are still needed.
Collapse
Affiliation(s)
- Linyu Wu
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Xiaohui Xian
- Department of Pathophysiology, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, 050051, Hebei, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050051, People's Republic of China
| | - Zixuan Tan
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, People's Republic of China
| | - Guangyu Xu
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, 050051, Hebei, People's Republic of China.
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050051, People's Republic of China.
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China.
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050051, People's Republic of China.
| |
Collapse
|
60
|
Hansson O, Blennow K, Zetterberg H, Dage J. Blood biomarkers for Alzheimer's disease in clinical practice and trials. NATURE AGING 2023; 3:506-519. [PMID: 37202517 PMCID: PMC10979350 DOI: 10.1038/s43587-023-00403-3] [Citation(s) in RCA: 156] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/21/2023] [Indexed: 05/20/2023]
Abstract
Blood-based biomarkers hold great promise to revolutionize the diagnostic and prognostic work-up of Alzheimer's disease (AD) in clinical practice. This is very timely, considering the recent development of anti-amyloid-β (Aβ) immunotherapies. Several assays for measuring phosphorylated tau (p-tau) in plasma exhibit high diagnostic accuracy in distinguishing AD from all other neurodegenerative diseases in patients with cognitive impairment. Prognostic models based on plasma p-tau levels can also predict future development of AD dementia in patients with mild cognitive complaints. The use of such high-performing plasma p-tau assays in the clinical practice of specialist memory clinics would reduce the need for more costly investigations involving cerebrospinal fluid samples or positron emission tomography. Indeed, blood-based biomarkers already facilitate identification of individuals with pre-symptomatic AD in the context of clinical trials. Longitudinal measurements of such biomarkers will also improve the detection of relevant disease-modifying effects of new drugs or lifestyle interventions.
Collapse
Affiliation(s)
- Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Lund, Sweden.
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for 27 Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Jeffrey Dage
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
61
|
Perez-Valero E, Gutierrez CAM, Lopez-Gordo MA, Alcalde SL. Evaluating the feasibility of cognitive impairment detection in Alzheimer's disease screening using a computerized visual dynamic test. J Neuroeng Rehabil 2023; 20:43. [PMID: 37046310 PMCID: PMC10091634 DOI: 10.1186/s12984-023-01155-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 03/06/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disease without known cure. However, early medical treatment can help control its progression and postpone intellectual decay. Since AD is preceded by a period of cognitive deterioration, the effective assessment of cognitive capabilities is crucial to develop reliable screening procedures. For this purpose, cognitive tests are extensively used to evaluate cognitive areas such as language, attention, or memory. METHODS In this work, we analyzed the potential of a visual dynamics evaluation, the rapid serial visual presentation task (RSVP), for the detection of cognitive impairment in AD. We compared this evaluation with two of the most extended brief cognitive tests applied in Spain: the Clock-drawing test (CDT) and the Phototest. For this purpose, we assessed a group of patients (mild AD and mild cognitive impairment) and controls, and we evaluated the ability of the three tests for the discrimination of the two groups. RESULTS The preliminary results obtained suggest the RSVP performance is statistically higher for the controls than for the patients (p-value = 0.013). Furthermore, we obtained promising classification results for this test (mean accuracy of 0.91 with 95% confidence interval 0.72, 0.97). CONCLUSIONS Since the RSVP is a computerized, auto-scored, and potentially self-administered brief test, it could contribute to speeding-up cognitive impairment screening and to reducing the associated costs. Furthermore, this evaluation could be combined with other tests to augment the efficiency of cognitive impairment screening protocols and to potentially monitor patients under medical treatment.
Collapse
Affiliation(s)
- Eduardo Perez-Valero
- Department of Computer Engineering, Automation and Robotics, University of Granada, Granada, Spain
- Brain-Computer Interfaces Laboratory, Research Centre for Information and Communications Technologies, Granada, Spain
| | - Christian A Morillas Gutierrez
- Department of Computer Engineering, Automation and Robotics, University of Granada, Granada, Spain
- Brain-Computer Interfaces Laboratory, Research Centre for Information and Communications Technologies, Granada, Spain
| | - Miguel Angel Lopez-Gordo
- Department of Signal Theory, Telematics, and Communications, University of Granada, Granada, Spain.
- Brain-Computer Interfaces Laboratory, Research Centre for Information and Communications Technologies, Granada, Spain.
| | | |
Collapse
|
62
|
Nir TM, Villalón-Reina JE, Salminen L, Haddad E, Zheng H, Thomopoulos SI, Jack CR, Weiner MW, Thompson PM, Jahanshad N. Cortical microstructural associations with CSF amyloid and pTau. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.10.23288366. [PMID: 37090601 PMCID: PMC10120803 DOI: 10.1101/2023.04.10.23288366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Diffusion MRI (dMRI) can be used to probe microstructural properties of brain tissue and holds great promise as a means to non-invasively map Alzheimer's disease (AD) pathology. Few studies have evaluated multi-shell dMRI models, such as neurite orientation dispersion and density imaging (NODDI) and mean apparent propagator (MAP)-MRI, in cortical gray matter where many of the earliest histopathological changes occur in AD. Here, we investigated the relationship between CSF pTau181 and Aβ1-42 burden and regional cortical NODDI and MAP-MRI indices in 46 cognitively unimpaired individuals, 18 with mild cognitive impairment, and two with dementia (mean age: 71.8±6.2 years) from the Alzheimer's Disease Neuroimaging Initiative. We compared findings to more conventional cortical thickness measures. Lower CSF Aβ1-42 and higher pTau181 were associated with cortical dMRI measures reflecting less hindered or restricted diffusion and greater diffusivity. Cortical dMRI measures were more widely associated with Aβ1-42 than pTau181 and better distinguished Aβ+ from Aβ- participants than pTau+/- participants. Conversely, cortical thickness was more tightly linked with pTau181. dMRI associations mediated the relationship between CSF markers and delayed logical memory performance, commonly impaired in early AD. dMRI measures sensitive to early AD pathogenesis and microstructural damage may elucidate mechanisms underlying cognitive decline.
Collapse
Affiliation(s)
- Talia M. Nir
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, United States
| | - Julio E. Villalón-Reina
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, United States
| | - Lauren Salminen
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, United States
| | - Elizabeth Haddad
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, United States
| | - Hong Zheng
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, United States
| | - Sophia I. Thomopoulos
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, United States
| | - Clifford R. Jack
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Michael W. Weiner
- Department of Radiology, School of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Paul M. Thompson
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, United States
| | - Neda Jahanshad
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, United States
| | | |
Collapse
|
63
|
Mieling M, Göttlich M, Yousuf M, Bunzeck N. Basal forebrain activity predicts functional degeneration in the entorhinal cortex and decreases with Alzheimer's Disease progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.28.534523. [PMID: 37034733 PMCID: PMC10081194 DOI: 10.1101/2023.03.28.534523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/19/2023]
Abstract
BACKGROUND AND OBJECTIVES Recent models of Alzheimer's Disease (AD) suggest the nucleus basalis of Meynert (NbM) as the origin of structural degeneration followed by the entorhinal cortex (EC). However, the functional properties of NbM and EC regarding amyloid-β and hyperphosphorylated tau remain unclear. METHODS We analyzed resting-state (rs)fMRI data with CSF assays from the Alzheimer's Disease Neuroimaging Initiative (ADNI, n=71) at baseline and two years later. RESULTS At baseline, local activity, as quantified by fractional amplitude of low-frequency fluctuations (fALFF), differentiated between normal and abnormal CSF groups in the NbM but not EC. Further, NbM activity linearly decreased as a function of CSF ratio, resembling the disease status. Finally, NbM activity predicted the annual percentage signal change in EC, but not the reverse, independent from CSF ratio. DISCUSSION Our findings give novel insights into the pathogenesis of AD by showing that local activity in NbM is affected by proteinopathology and predicts functional degeneration within the EC.
Collapse
Affiliation(s)
- Marthe Mieling
- Department of Psychology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Martin Göttlich
- Department of Neurology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
- Center of Brain, Behavior and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Mushfa Yousuf
- Department of Psychology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Nico Bunzeck
- Department of Psychology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
- Center of Brain, Behavior and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | | |
Collapse
|
64
|
Alban SL, Lynch KM, Ringman JM, Toga AW, Chui HC, Sepehrband F, Choupan J. The association between white matter hyperintensities and amyloid and tau deposition. Neuroimage Clin 2023; 38:103383. [PMID: 36965457 PMCID: PMC10060905 DOI: 10.1016/j.nicl.2023.103383] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 02/09/2023] [Accepted: 03/16/2023] [Indexed: 03/22/2023]
Abstract
White matter hyperintensities (WMHs) frequently occur in Alzheimer's Disease (AD) and have a contribution from ischemia, though their relationship with β-amyloid and cardiovascular risk factors (CVRFs) is not completely understood. We used AT classification to categorize individuals based on their β-amyloid and tau pathologies, then assessed the effects of β-amyloid and tau on WMH volume and number. We then determined regions in which β-amyloid and WMH accumulation were related. Last, we analyzed the effects of various CVRFs on WMHs. As secondary analyses, we observed effects of age and sex differences, atrophy, cognitive scores, and APOE genotype. PET, MRI, FLAIR, demographic, and cardiovascular health data was collected from the Alzheimer's Disease Neuroimaging Initiative (ADNI-3) (N = 287, 48 % male). Participants were categorized as A + and T + if their Florbetapir SUVR and Flortaucipir SUVR were above 0.79 and 1.25, respectively. WMHs were mapped on MRI using a deep convolutional neural network (Sepehrband et al., 2020). CVRF scores were based on history of hypertension, systolic and diastolic blood pressure, pulse rate, respiration rate, BMI, and a cumulative score with 6 being the maximum score. Regression models and Pearson correlations were used to test associations and correlations between variables, respectively, with age, sex, years of education, and scanner manufacturer as covariates of no interest. WMH volume percent was significantly associated with global β-amyloid (r = 0.28, p < 0.001), but not tau (r = 0.05, p = 0.25). WMH volume percent was higher in individuals with either A + or T + pathology compared to controls, particularly within in the A+/T + group (p = 0.007, Cohen's d = 0.4, t = -2.5). Individual CVRFs nor cumulative CVRF scores were associated with increased WMH volume. Finally, the regions where β-amyloid and WMH count were most positively associated were the middle temporal region in the right hemisphere (r = 0.18, p = 0.002) and the fusiform region in the left hemisphere (r = 0.017, p = 0.005). β-amyloid and WMH have a clear association, though the mechanism facilitating this association is still not fully understood. The associations found between β-amyloid and WMH burden emphasizes the relationship between β-amyloid and vascular lesion formation while factors like CVRFs, age, and sex affect AD development through various mechanisms. These findings highlight potential causes and mechanisms of AD as targets for future preventions and treatments. Going forward, a larger emphasis may be placed on β-amyloid's vascular effects and the implications of impaired brain clearance in AD.
Collapse
Affiliation(s)
- Sierra L Alban
- Laboratory of NeuroImaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kirsten M Lynch
- Laboratory of NeuroImaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - John M Ringman
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Arthur W Toga
- Laboratory of NeuroImaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Helena C Chui
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Farshid Sepehrband
- Laboratory of NeuroImaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jeiran Choupan
- Laboratory of NeuroImaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; NeuroScope Inc., Scarsdale, NY, USA
| |
Collapse
|
65
|
Role of Tau in Various Tauopathies, Treatment Approaches, and Emerging Role of Nanotechnology in Neurodegenerative Disorders. Mol Neurobiol 2023; 60:1690-1720. [PMID: 36562884 DOI: 10.1007/s12035-022-03164-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
A few protein kinases and phosphatases regulate tau protein phosphorylation and an imbalance in their enzyme activity results in tau hyper-phosphorylation. Aberrant tau phosphorylation causes tau to dissociate from the microtubules and clump together in the cytosol to form neurofibrillary tangles (NFTs), which lead to the progression of neurodegenerative disorders including Alzheimer's disease (AD) and other tauopathies. Hence, targeting hyperphosphorylated tau protein is a restorative approach for treating neurodegenerative tauopathies. The cyclin-dependent kinase (Cdk5) and the glycogen synthase kinase (GSK3β) have both been implicated in aberrant tau hyperphosphorylation. The limited transport of drugs through the blood-brain barrier (BBB) for reaching the central nervous system (CNS) thus represents a significant problem in the development of drugs. Drug delivery systems based on nanocarriers help solve this problem. In this review, we discuss the tau protein, regulation of tau phosphorylation and abnormal hyperphosphorylation, drugs in use or under clinical trials, and treatment strategies for tauopathies based on the critical role of tau hyperphosphorylation in the pathogenesis of the disease. Pathology of neurodegenerative disease due to hyperphosphorylation and various therapeutic approaches including nanotechnology for its treatment.
Collapse
|
66
|
Constantinides VC, Paraskevas GP, Boufidou F, Bourbouli M, Pyrgelis ES, Stefanis L, Kapaki E. CSF Aβ42 and Aβ42/Aβ40 Ratio in Alzheimer's Disease and Frontotemporal Dementias. Diagnostics (Basel) 2023; 13:diagnostics13040783. [PMID: 36832271 PMCID: PMC9955886 DOI: 10.3390/diagnostics13040783] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Alzheimer's disease dementia (ADD) may manifest with atypical phenotypes, resembling behavioral variant frontotemporal dementia (bvFTD) and corticobasal syndrome (CBS), phenotypes which typically have an underlying frontotemporal lobar degeneration with tau proteinopathy (FTLD-tau), such as Pick's disease, corticobasal degeneration (CBD), progressive supranuclear palsy (PSP), or FTLD with TDP-43 proteinopathy (FTLD-TDP). CSF biomarkers total and phosphorylated tau (τT and τP-181), and amyloid beta with 42 and 40 amino acids (Aβ42 and Aβ40) are biomarkers of AD pathology. The primary aim of this study was to compare the diagnostic accuracy of Aβ42 to Aβ42/Aβ40 ratio in: (a) differentiating ADD vs. frontotemporal dementias; (b) patients with AD pathology vs. non-AD pathologies; (c) compare biomarker ratios and composite markers to single CSF biomarkers in the differentiation of AD from FTD; Methods: In total, 263 subjects were included (ADD: n = 98; bvFTD: n = 49; PSP: n = 50; CBD: n = 45; controls: n = 21). CSF biomarkers were measured by commercially available ELISAs (EUROIMMUN). Multiple biomarker ratios (Aβ42/Aβ40; τT/τP-181; τT/Aβ42; τP-181/Aβ42) and composite markers (t-tau: τT/(Aβ42/Aβ40); p-tau: τP-181/(Aβ42/Aβ40) were calculated. ROC curve analysis was performed to compare AUCs of Aβ42 and Aβ42/Aβ40 ratio and relevant composite markers between ADD and FTD, as defined clinically. BIOMARKAPD/ABSI criteria (abnormal τT, τP-181 Aβ42, and Aβ42/Aβ40 ratio) were used to re-classify all patients into AD pathology vs. non-AD pathologies, and ROC curve analysis was repeated to compare Aβ42 and Aβ42/Aβ40; Results: Aβ42 did not differ from Aβ42/Aβ40 ratio in the differentiation of ADD from FTD (AUCs 0.752 and 0.788 respectively; p = 0.212). The τT/Aβ42 ratio provided maximal discrimination between ADD and FTD (AUC:0.893; sensitivity 88.8%, specificity 80%). BIOMARKAPD/ABSI criteria classified 60 patients as having AD pathology and 211 as non-AD. A total of 22 had discrepant results and were excluded. Aβ42/Aβ40 ratio was superior to Aβ42 in the differentiation of AD pathology from non-AD pathology (AUCs: 0.939 and 0.831, respectively; p < 0.001). In general, biomarker ratios and composite markers were superior to single CSF biomarkers in both analyses. CONCLUSIONS Aβ42/Aβ40 ratio is superior to Aβ42 in identifying AD pathology, irrespective of the clinical phenotype. CSF biomarker ratios and composite markers provide higher diagnostic accuracy compared to single CSF biomarkers.
Collapse
Affiliation(s)
- Vasilios C. Constantinides
- First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
- Correspondence: ; Tel.: +30-21-0728-9285
| | - George P. Paraskevas
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
- Second Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, Rimini 1, 12462 Athens, Greece
| | - Fotini Boufidou
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| | - Mara Bourbouli
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| | - Efstratios-Stylianos Pyrgelis
- First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| | - Leonidas Stefanis
- First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| | - Elisabeth Kapaki
- First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| |
Collapse
|
67
|
Sun Y, Zhao Y, Hu K, Wang M, Liu Y, Liu B. Distinct spatiotemporal subtypes of amyloid deposition are associated with diverging disease profiles in cognitively normal and mild cognitive impairment individuals. Transl Psychiatry 2023; 13:35. [PMID: 36732496 PMCID: PMC9895066 DOI: 10.1038/s41398-023-02328-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/04/2023] [Accepted: 01/19/2023] [Indexed: 02/04/2023] Open
Abstract
We aimed to investigate the relationship between spatiotemporal changes of amyloid deposition and Alzheimer's disease (AD) profiles in cognitively normal (CN) and those with mild cognitive impairment (MCI). Using a data-driven method and amyloid-PET data, we identified and validated two subtypes in two independent datasets (discovery dataset: N = 548, age = 72.4 ± 6.78, 49% female; validation dataset: N = 348, age = 74.9 ± 8.16, 47% female) from the Alzheimer's Disease Neuroimaging Initiative across a range of individuals who were CN or had MCI. The two subtypes showed distinct regional progression patterns and presented distinct genetic, clinical and biomarker characteristics. The cortex-priority subtype was more likely to show typical clinical syndromes of symptomatic AD and vice versa. Furthermore, the regional progression patterns were associated with clinical and biomarker profiles. In sum, our findings suggest that the spatiotemporal variants of amyloid depositions are in close association with disease trajectories; these findings may provide insight into the disease monitoring and enrollment of therapeutic trials in AD.
Collapse
Affiliation(s)
- Yuqing Sun
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yuxin Zhao
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Ke Hu
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Meng Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yong Liu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, 100876, China.
| | - Bing Liu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China.
- Chinese Institute for Brain Research, Beijing, 102206, China.
| |
Collapse
|
68
|
Loftus JR, Puri S, Meyers SP. Multimodality imaging of neurodegenerative disorders with a focus on multiparametric magnetic resonance and molecular imaging. Insights Imaging 2023; 14:8. [PMID: 36645560 PMCID: PMC9842851 DOI: 10.1186/s13244-022-01358-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 12/13/2022] [Indexed: 01/17/2023] Open
Abstract
Neurodegenerative diseases afflict a large number of persons worldwide, with the prevalence and incidence of dementia rapidly increasing. Despite their prevalence, clinical diagnosis of dementia syndromes remains imperfect with limited specificity. Conventional structural-based imaging techniques also lack the accuracy necessary for confident diagnosis. Multiparametric magnetic resonance imaging and molecular imaging provide the promise of improving specificity and sensitivity in the diagnosis of neurodegenerative disease as well as therapeutic monitoring of monoclonal antibody therapy. This educational review will briefly focus on the epidemiology, clinical presentation, and pathologic findings of common and uncommon neurodegenerative diseases. Imaging features of each disease spanning from conventional magnetic resonance sequences to advanced multiparametric methods such as resting-state functional magnetic resonance imaging and arterial spin labeling imaging will be described in detail. Additionally, the review will explore the findings of each diagnosis on molecular imaging including single-photon emission computed tomography and positron emission tomography with a variety of clinically used and experimental radiotracers. The literature and clinical cases provided demonstrate the power of advanced magnetic resonance imaging and molecular techniques in the diagnosis of neurodegenerative diseases and areas of future and ongoing research. With the advent of combined positron emission tomography/magnetic resonance imaging scanners, hybrid protocols utilizing both techniques are an attractive option for improving the evaluation of neurodegenerative diseases.
Collapse
Affiliation(s)
- James Ryan Loftus
- grid.412750.50000 0004 1936 9166Department of Imaging Sciences, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14642 USA
| | - Savita Puri
- grid.412750.50000 0004 1936 9166Department of Imaging Sciences, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14642 USA
| | - Steven P. Meyers
- grid.412750.50000 0004 1936 9166Department of Imaging Sciences, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14642 USA
| |
Collapse
|
69
|
Abbate C, Trimarchi PD, Fumagalli GG, Gallucci A, Tomasini E, Fracchia S, Rebecchi I, Morello E, Fontanella A, Parisi PM, Tartarone F, Giunco F, Ciccone S, Nicolini P, Lucchi T, Arosio B, Inglese S, Rossi PD. Diencephalic versus Hippocampal Amnesia in Alzheimer's Disease: The Possible Confabulation-Misidentification Phenotype. J Alzheimers Dis 2023; 91:363-388. [PMID: 36442200 PMCID: PMC9881034 DOI: 10.3233/jad-220919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is clinically heterogeneous, including the classical-amnesic (CA-) phenotype and some variants. OBJECTIVE We aim to describe a further presentation we (re)named confabulation-misidentification (CM-) phenotype. METHODS We performed a retrospective longitudinal case-series study of 17 AD outpatients with the possible CM-phenotype (CM-ADs). Then, in a cross-sectional study, we compared the CM-ADs to a sample of 30 AD patients with the CA-phenotype (CA-ADs). The primary outcome was the frequency of cognitive and behavioral features. Data were analyzed as differences in percentage by non-parametric Chi Square and mean differences by parametric T-test. RESULTS Anterograde amnesia (100%) with early confabulation (88.2%), disorientation (88.2%) and non-infrequently retrograde amnesia (64.7%) associated with reduced insight (88.2%), moderate prefrontal executive impairment (94.1%) and attention deficits (82.3%) dominated the CM-phenotype. Neuropsychiatric features with striking misidentification (52.9%), other less-structured delusions (70.6%), and brief hallucinations (64.7%) were present. Marked behavioral disturbances were present early in some patients and very common at later stages. At the baseline, the CM-ADs showed more confabulation (p < 0.001), temporal disorientation (p < 0.02), misidentification (p = 0.013), other delusions (p = 0.002), and logorrhea (p = 0.004) than the CA-ADs. In addition, more social disinhibition (p = 0.018), reduction of insight (p = 0.029), and hallucination (p = 0.03) persisted at 12 months from baseline. Both the CA- and CM-ADs showed anterior and medial temporal atrophy. Compared to HCs, the CM-ADs showed more right fronto-insular atrophy, while the CA-ADs showed more dorsal parietal, precuneus, and right parietal atrophy. CONCLUSION We described an AD phenotype resembling diencephalic rather than hippocampal amnesia and overlapping the past-century description of presbyophrenia.
Collapse
Affiliation(s)
- Carlo Abbate
- Istituto Palazzolo, IRCCS Fondazione Don Carlo Gnocchi Onlus, Milan, Italy
| | | | - Giorgio G. Fumagalli
- Neurology Unit, IRCCS Fondazione Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Alessia Gallucci
- Istituto Palazzolo, IRCCS Fondazione Don Carlo Gnocchi Onlus, Milan, Italy
- Ph.D. Program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Emanuele Tomasini
- Istituto Palazzolo, IRCCS Fondazione Don Carlo Gnocchi Onlus, Milan, Italy
| | - Stefania Fracchia
- Istituto Palazzolo, IRCCS Fondazione Don Carlo Gnocchi Onlus, Milan, Italy
| | - Isabella Rebecchi
- Istituto Palazzolo, IRCCS Fondazione Don Carlo Gnocchi Onlus, Milan, Italy
| | - Elisabetta Morello
- Istituto Palazzolo, IRCCS Fondazione Don Carlo Gnocchi Onlus, Milan, Italy
| | - Anna Fontanella
- Istituto Palazzolo, IRCCS Fondazione Don Carlo Gnocchi Onlus, Milan, Italy
| | - Paola M.R. Parisi
- Istituto Palazzolo, IRCCS Fondazione Don Carlo Gnocchi Onlus, Milan, Italy
| | - Federica Tartarone
- Istituto Palazzolo, IRCCS Fondazione Don Carlo Gnocchi Onlus, Milan, Italy
| | - Fabrizio Giunco
- Istituto Palazzolo, IRCCS Fondazione Don Carlo Gnocchi Onlus, Milan, Italy
| | - Simona Ciccone
- Geriatric Unit, IRCCS Fondazione Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Paola Nicolini
- Geriatric Unit, IRCCS Fondazione Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Tiziano Lucchi
- Geriatric Unit, IRCCS Fondazione Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Beatrice Arosio
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Silvia Inglese
- Geriatric Unit, IRCCS Fondazione Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Paolo D. Rossi
- Geriatric Unit, IRCCS Fondazione Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
70
|
Sangalli L, Boggero IA. The impact of sleep components, quality and patterns on glymphatic system functioning in healthy adults: A systematic review. Sleep Med 2023; 101:322-349. [PMID: 36481512 DOI: 10.1016/j.sleep.2022.11.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/04/2022] [Accepted: 11/13/2022] [Indexed: 11/24/2022]
Abstract
OBJECTIVE The glymphatic system is thought to be responsible for waste clearance in the brain. As it is primarily active during sleep, different components of sleep, subjective sleep quality, and sleep patterns may contribute to glymphatic functioning. This systematic review aimed at exploring the effect of sleep components, sleep quality, and sleep patterns on outcomes associated with the glymphatic system in healthy adults. METHODS PubMed®, Scopus, and Web of Science were searched for studies published in English until December 2021. Articles subjectively or objectively investigating sleep components (total sleep time, time in bed, sleep efficiency, sleep onset latency, wake-up after sleep onset, sleep stage, awakenings), sleep quality, or sleep pattern in healthy individuals, on outcomes associated with glymphatic system (levels of amyloid-β, tau, α-synuclein; cerebrospinal fluid, perivascular spaces; apolipoprotein E) were selected. RESULTS Out of 8359 records screened, 51 studies were included. Overall, contradictory findings were observed according to different sleep assessment method. The most frequently assessed sleep parameters were total sleep time, sleep quality, and sleep efficiency. No association was found between sleep efficiency and amyloid-β, and between slow-wave activity and tau. Most of the studies did not find any correlation between total sleep time and amyloid-β nor tau level. Opposing results correlated sleep quality with amyloid-β and tau. CONCLUSIONS This review highlighted inconsistent results across the studies; as such, the specific association between the glymphatic system and sleep parameters in healthy adults remains poorly understood. Due to the heterogeneity of sleep assessment methods and the self-reported data representing the majority of the observations, future studies with universal study design and sleep methodology in healthy individuals are advocated.
Collapse
Affiliation(s)
- L Sangalli
- Department of Oral Health Science, Division of Orofacial Pain, University of Kentucky, College of Dentistry, Lexington, Kentucky, USA; College of Dental Medicine - Illinois, Downers Grove, Illinois, USA.
| | - I A Boggero
- Department of Oral Health Science, Division of Orofacial Pain, University of Kentucky, College of Dentistry, Lexington, Kentucky, USA; Department of Psychology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
71
|
Shah AJ, Mohi-Ud-Din R, Sabreen S, Wani TU, Jan R, Javed MN, Mir PA, Mir RH, Masoodi MH. Clinical Biomarkers and Novel Drug Targets to Cut Gordian Knots of Alzheimer's Disease. Curr Mol Pharmacol 2023; 16:254-279. [PMID: 36056834 DOI: 10.2174/1874467215666220903095837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 12/03/2021] [Accepted: 12/16/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Alzheimer's disease (AD), the primary cause of dementia, escalating worldwide, has no proper diagnosis or effective treatment. Neuronal cell death and impairment of cognitive abilities, possibly triggered by several brain mechanisms, are the most significant characteristic of this disorder. METHODS A multitude of pharmacological targets have been identified for potential drug design against AD. Although many advances in treatment strategies have been made to correct various abnormalities, these often exhibit limited clinical significance because this disease aggressively progresses into different regions of the brain, causing severe deterioration. RESULTS These biomarkers can be game-changers for early detection and timely monitoring of such disorders. CONCLUSION This review covers clinically significant biomarkers of AD for precise and early monitoring of risk factors and stages of this disease, the potential site of action and novel targets for drugs, and pharmacological approaches to clinical management.
Collapse
Affiliation(s)
- Abdul Jalil Shah
- Department of Pharmaceutical Sciences, Pharmaceutical Chemistry Division, University of Kashmir, Hazratbal, Srinagar- 190006, Kashmir, India
| | - Roohi Mohi-Ud-Din
- Department of General Medicine, Sher-I-Kashmir Institute of Medical Sciences (SKIMS), Srinagar- 190011, Jammu and Kashmir, India
| | - Saba Sabreen
- Department of Pharmaceutical Sciences, Pharmaceutical Chemistry Division, University of Kashmir, Hazratbal, Srinagar- 190006, Kashmir, India
| | - Taha Umair Wani
- Department of Pharmaceutical Sciences, Pharmaceutics Lab, School of Applied Sciences and Technology, University of Kashmir, Hazratbal, Srinagar-190006, Kashmir India
| | - Rafia Jan
- Defence Research and Development Organization (DRDO), Hospital, Khonmoh, Srinagar 190001, Jammu & Kashmir, India
| | - Md Noushad Javed
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmaceutics, KR Mangalam University, Gurugram, India
| | - Prince Ahad Mir
- Khalsa College of Pharmacy, G.T. Road, Amritsar-143002, Punjab, India
| | - Reyaz Hassan Mir
- Department of Pharmaceutical Sciences, Pharmaceutical Chemistry Division, University of Kashmir, Hazratbal, Srinagar- 190006, Kashmir, India
- Pharmaceutical Chemistry Division, Chandigarh College of Pharmacy, Mohali, Punjab 140307, India
| | - Mubashir Hussain Masoodi
- Department of Pharmaceutical Sciences, Pharmaceutical Chemistry Division, University of Kashmir, Hazratbal, Srinagar- 190006, Kashmir, India
| |
Collapse
|
72
|
Tarawneh R, Kasper RS, Sanford J, Phuah C, Hassenstab J, Cruchaga C. Vascular endothelial-cadherin as a marker of endothelial injury in preclinical Alzheimer disease. Ann Clin Transl Neurol 2022; 9:1926-1940. [PMID: 36342663 PMCID: PMC9735377 DOI: 10.1002/acn3.51685] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/02/2022] [Accepted: 10/10/2022] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVE Endothelial dysfunction is an early and prevalent pathology in Alzheimer disease (AD). We here investigate the value of vascular endothelial-cadherin (VEC) as a cerebrospinal fluid (CSF) marker of endothelial injury in preclinical AD. METHODS Cognitively normal participants (Clinical Dementia Rating [CDR] 0) from the Knight Washington University-ADRC were included in this study (n = 700). Preclinical Alzheimer's Cognitive Composite (PACC) scores, CSF VEC, tau, p-tau181, Aβ42/Aβ40, neurofilament light-chain (NFL) levels, and magnetic resonance imaging (MRI) assessments of white matter injury (WMI) were obtained from all participants. A subset of participants underwent brain amyloid imaging using positron emission tomography (amyloid-PET) (n = 534). Linear regression examined associations of CSF VEC with PACC and individual cognitive scores in preclinical AD. Mediation analyses examined whether CSF VEC mediated effects of CSF amyloid and tau markers on cognition in preclinical AD. RESULTS CSF VEC levels significantly correlated with PACC and individual cognitive scores in participants with amyloid (A+T±N±; n = 558) or those with amyloid and tau pathologies (A+T+N±; n = 259), after adjusting for covariates. CSF VEC also correlated with CSF measures of amyloid, tau, and neurodegeneration and global amyloid burden on amyloid-PET scans in our cohort. Importantly, our findings suggest that CSF VEC mediates associations of CSF Aβ42/Aβ40, p-tau181, and global amyloid burden with cognitive outcomes in preclinical AD. INTERPRETATION Our results support the utility of CSF VEC as a marker of endothelial injury in AD and highlight the importance of endothelial injury as an early pathology that contributes to cognitive impairment in even the earliest preclinical stages.
Collapse
Affiliation(s)
- Rawan Tarawneh
- Department of NeurologyUniversity of New MexicoAlbuquerqueNew MexicoUSA
- Center for Memory and AgingUniversity of New MexicoAlbuquerqueNew MexicoUSA
| | - Rachel S. Kasper
- Department of NeurologyUniversity of New MexicoAlbuquerqueNew MexicoUSA
| | - Jessie Sanford
- Department of PsychiatryWashington University in St LouisSt. LouisMissouriUSA
- NeuroGenomics and Informatics CenterWashington University in St LouisMissouriUSA
| | - Chia‐Ling Phuah
- NeuroGenomics and Informatics CenterWashington University in St LouisMissouriUSA
- Department of NeurologyWashington University in St LouisSt. LouisMissouriUSA
| | - Jason Hassenstab
- Department of PsychologyWashington University in St LouisSt. LouisMissouriUSA
| | - Carlos Cruchaga
- Department of PsychiatryWashington University in St LouisSt. LouisMissouriUSA
- NeuroGenomics and Informatics CenterWashington University in St LouisMissouriUSA
| |
Collapse
|
73
|
Zhang Y, Ghose U, Buckley NJ, Engelborghs S, Sleegers K, Frisoni GB, Wallin A, Lleó A, Popp J, Martinez-Lage P, Legido-Quigley C, Barkhof F, Zetterberg H, Visser PJ, Bertram L, Lovestone S, Nevado-Holgado AJ, Shi L. Predicting AT(N) pathologies in Alzheimer's disease from blood-based proteomic data using neural networks. Front Aging Neurosci 2022; 14:1040001. [PMID: 36523958 PMCID: PMC9746615 DOI: 10.3389/fnagi.2022.1040001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/04/2022] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Blood-based biomarkers represent a promising approach to help identify early Alzheimer's disease (AD). Previous research has applied traditional machine learning (ML) to analyze plasma omics data and search for potential biomarkers, but the most modern ML methods based on deep learning has however been scarcely explored. In the current study, we aim to harness the power of state-of-the-art deep learning neural networks (NNs) to identify plasma proteins that predict amyloid, tau, and neurodegeneration (AT[N]) pathologies in AD. METHODS We measured 3,635 proteins using SOMAscan in 881 participants from the European Medical Information Framework for AD Multimodal Biomarker Discovery study (EMIF-AD MBD). Participants underwent measurements of brain amyloid β (Aβ) burden, phosphorylated tau (p-tau) burden, and total tau (t-tau) burden to determine their AT(N) statuses. We ranked proteins by their association with Aβ, p-tau, t-tau, and AT(N), and fed the top 100 proteins along with age and apolipoprotein E (APOE) status into NN classifiers as input features to predict these four outcomes relevant to AD. We compared NN performance of using proteins, age, and APOE genotype with performance of using age and APOE status alone to identify protein panels that optimally improved the prediction over these main risk factors. Proteins that improved the prediction for each outcome were aggregated and nominated for pathway enrichment and protein-protein interaction enrichment analysis. RESULTS Age and APOE alone predicted Aβ, p-tau, t-tau, and AT(N) burden with area under the curve (AUC) scores of 0.748, 0.662, 0.710, and 0.795. The addition of proteins significantly improved AUCs to 0.782, 0.674, 0.734, and 0.831, respectively. The identified proteins were enriched in five clusters of AD-associated pathways including human immunodeficiency virus 1 infection, p53 signaling pathway, and phosphoinositide-3-kinase-protein kinase B/Akt signaling pathway. CONCLUSION Combined with age and APOE genotype, the proteins identified have the potential to serve as blood-based biomarkers for AD and await validation in future studies. While the NNs did not achieve better scores than the support vector machine model used in our previous study, their performances were likely limited by small sample size.
Collapse
Affiliation(s)
- Yuting Zhang
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Upamanyu Ghose
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Noel J. Buckley
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Sebastiaan Engelborghs
- Department of Biomedical Sciences, Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Department of Neurology, Universitair Ziekenhuis Brussel, Brussels, Belgium
- Center for Neurociences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | - Kristel Sleegers
- Complex Genetics Group, VIB Center for Molecular Neurology, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Anders Wallin
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Alberto Lleó
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Julius Popp
- Department of Psychiatry, University Hospital of Lausanne, Lausanne, Switzerland
- Department of Geriatric Psychiatry, University Hospital of Psychiatry and University of Zürich, Zürich, Switzerland
| | | | - Cristina Legido-Quigley
- Kings College London, London, United Kingdom
- The Systems Medicine Group, Steno Diabetes Center, Gentofte, Denmark
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, Netherlands
- University College London (UCL) Institutes of Neurology and Healthcare Engineering, London, United Kingdom
| | - 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
- UK Dementia Research Institute at UCL, London, United Kingdom
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, Netherlands
- Alzheimer Center, VU University Medical Center, Amsterdam, Netherlands
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, University of Lübeck, Lübeck, Germany
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Simon Lovestone
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Janssen R&D, High Wycombe, United Kingdom
| | | | - Liu Shi
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
74
|
Cistaro A, Quartuccio N, Cassalia L, Vai D, Guerra UP, Atzori C, Rainero I, Imperiale D. Brain 18 F-Florbetapir PET/CT Findings in an Early-onset Alzheimer Disease Patient Carrying Presenilin-1 G378E Mutation. Alzheimer Dis Assoc Disord 2022; 36:347-349. [PMID: 34132671 DOI: 10.1097/wad.0000000000000461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023]
Abstract
Positron emission tomography (PET) with 18 F-Fluorodeoxyglucose ( 18 F-FDG) plays an outstanding role in the diagnostic work-up of dementia. Amyloid PET imaging is a complementary imaging technique for the early detection of Alzheimer disease (AD). β-amyloid precursor protein ( APP ), Presenilin-1 ( PSEN1 ) and Presenilin-2 ( PSEN2 ) are the 3 main causative genes responsible for autosomal dominant early-onset Alzheimer disease (EOAD). This is the first report of 18 F-Florbetapir amyloid imaging findings in a 35-year-old male patient with EOAD carrying the G378E mutation in PSEN1 gene. Brain computed tomography (CT) and magnetic resonance imaging scans showed remarkable cerebral atrophy with dilatation of the cerebrospinal fluid spaces; furthermore, a 18 F-Florbetapir PET/CT scan demonstrated also widespread remarkable accumulation of the amyloid tracer in the cerebral cortex, with reduction of the normal contrast between white and gray matter and flattening of the external cortical margins. Furthermore, PET/CT showed intense 18 F-florbetapir uptake in the striatum and in the thalamus bilaterally. Our case supports the usefulness of amyloid PET imaging in the diagnostic work-up of EOAD.
Collapse
Affiliation(s)
- Angelina Cistaro
- Nuclear Medicine Department, Ospedali Galliera, Genoa
- AIMN Neuroimaging Study Group, Milan
| | - Natale Quartuccio
- AIMN Neuroimaging Study Group, Milan
- Nuclear Medicine Unit, A.R.N.A.S. Ospedali Civico, Di Cristina e Benfratelli, Palermo
| | - Laura Cassalia
- Department of Radiology, Institute of Radiology, "Magna Grecia" University, Catanzaro
| | - Daniela Vai
- Neurology Unit and Human TSE Regional Center, "Amedeo di Savoia" & "Maria Vittoria" Hospital, Turin
| | | | - Cristiana Atzori
- Neurology Unit and Human TSE Regional Center, "Amedeo di Savoia" & "Maria Vittoria" Hospital, Turin
| | - Innocenzo Rainero
- Neurology I, Department of Neuroscience "Rita Levi Montalcini," University of Torino, Torino, Italy
| | - Daniele Imperiale
- Neurology Unit and Human TSE Regional Center, "Amedeo di Savoia" & "Maria Vittoria" Hospital, Turin
| |
Collapse
|
75
|
Xu C, Zhao L, Dong C. A Review of Application of Aβ42/40 Ratio in Diagnosis and Prognosis of Alzheimer’s Disease. J Alzheimers Dis 2022; 90:495-512. [DOI: 10.3233/jad-220673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The number of patients with Alzheimer’s disease (AD) and non-Alzheimer’s disease (non-AD) has drastically increased over recent decades. The amyloid cascade hypothesis attributes a vital role to amyloid-β protein (Aβ) in the pathogenesis of AD. As the main pathological hallmark of AD, amyloid plaques consist of merely the 42 and 40 amino acid variants of Aβ (Aβ 42 and Aβ 40). The cerebrospinal fluid (CSF) biomarker Aβ 42/40 has been extensively investigated and eventually integrated into important diagnostic tools to support the clinical diagnosis of AD. With the development of highly sensitive assays and technologies, blood-based Aβ 42/40, which was obtained using a minimally invasive and cost-effective method, has been proven to be abnormal in synchrony with CSF biomarker values. This paper presents the recent progress of the CSF Aβ 42/40 ratio and plasma Aβ 42/40 for AD as well as their potential clinical application as diagnostic markers or screening tools for dementia.
Collapse
Affiliation(s)
- Chang Xu
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Li Zhao
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Chunbo Dong
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
76
|
Guo X, Chen K, Chen Y, Xiong C, Su Y, Yao L, Reiman EM. A Computational Monte Carlo Simulation Strategy to Determine the Temporal Ordering of Abnormal Age Onset Among Biomarkers of Alzheimer's Disease. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:2613-2622. [PMID: 34428151 PMCID: PMC9588284 DOI: 10.1109/tcbb.2021.3106939] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
To quantitatively determining the temporal ordering of abnormal age onsets (AAO) among various biomarkers for Alzheimer's disease (AD), we introduced a computational Monte-Carlo simulation (CMCS) to statistically examine such ordering of an AAO pair or over all AAOs. The CMCS 1) simulates longitudinal data, estimates AAO for each iteration, and finally assesses the type-I error of an AAO pair or all AAO ordering. Using hippocampus volume (VHC), cerebral glucose hypometabolic convergence index (HCI), plasma neurofilament light (NfL), mini-mental state exam (MMSE), the auditory verbal learning test-long term memory (AVLT-LTM), short term memory (AVLT-STM) and clinical-dementia rating sum of box scale (CDR-SOB) from 382 mild cognitive impairment converters and non-converters, the CMCS estimated type-I error for the earlier AAO of VHC, AVLT_STM and AVLT_LTM each than MMSE was significant (p<0.002). The type-I error for the overall AAO temporal ordering of VHC ≤ AVLT_STM ≤ AVLT_LTM < HCI ≤ MMSE ≤ CDR-SOB ≤ NfL was p = 0.012. These findings showed that our CMCS is capable of providing statistical inferences for quantifying AAO ordering which has important implications in advancing our understanding of AD.
Collapse
|
77
|
Moguilner S, Birba A, Fittipaldi S, Gonzalez-Campo C, Tagliazucchi E, Reyes P, Matallana D, Parra MA, Slachevsky A, Farías G, Cruzat J, García A, Eyre HA, Joie RL, Rabinovici G, Whelan R, Ibáñez A. Multi-feature computational framework for combined signatures of dementia in underrepresented settings. J Neural Eng 2022; 19:10.1088/1741-2552/ac87d0. [PMID: 35940105 PMCID: PMC11177279 DOI: 10.1088/1741-2552/ac87d0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/08/2022] [Indexed: 11/11/2022]
Abstract
Objective.The differential diagnosis of behavioral variant frontotemporal dementia (bvFTD) and Alzheimer's disease (AD) remains challenging in underrepresented, underdiagnosed groups, including Latinos, as advanced biomarkers are rarely available. Recent guidelines for the study of dementia highlight the critical role of biomarkers. Thus, novel cost-effective complementary approaches are required in clinical settings.Approach. We developed a novel framework based on a gradient boosting machine learning classifier, tuned by Bayesian optimization, on a multi-feature multimodal approach (combining demographic, neuropsychological, magnetic resonance imaging (MRI), and electroencephalography/functional MRI connectivity data) to characterize neurodegeneration using site harmonization and sequential feature selection. We assessed 54 bvFTD and 76 AD patients and 152 healthy controls (HCs) from a Latin American consortium (ReDLat).Main results. The multimodal model yielded high area under the curve classification values (bvFTD patients vs HCs: 0.93 (±0.01); AD patients vs HCs: 0.95 (±0.01); bvFTD vs AD patients: 0.92 (±0.01)). The feature selection approach successfully filtered non-informative multimodal markers (from thousands to dozens).Results. Proved robust against multimodal heterogeneity, sociodemographic variability, and missing data.Significance. The model accurately identified dementia subtypes using measures readily available in underrepresented settings, with a similar performance than advanced biomarkers. This approach, if confirmed and replicated, may potentially complement clinical assessments in developing countries.
Collapse
Affiliation(s)
- Sebastian Moguilner
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), CA, United States of America
- Cognitive Neuroscience Center (CNC), Universidad de San Andrés, Buenos Aires, Argentina
- Latin American Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile
- Trinity College Dublin, Dublin, Ireland
| | - Agustina Birba
- Cognitive Neuroscience Center (CNC), Universidad de San Andrés, Buenos Aires, Argentina
- Latin American Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Sol Fittipaldi
- Cognitive Neuroscience Center (CNC), Universidad de San Andrés, Buenos Aires, Argentina
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | | | - Enzo Tagliazucchi
- Latin American Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- Department of Physics, University of Buenos Aires, Buenos Aires, Argentina
| | - Pablo Reyes
- Medical School, Aging Institute, Psychiatry and Mental Health, Pontificia Universidad Javeriana, Bogota, Colombia
| | - Diana Matallana
- Medical School, Aging Institute, Psychiatry and Mental Health, Pontificia Universidad Javeriana, Bogota, Colombia
| | - Mario A Parra
- MAP: School of Psychological Sciences and Health, University of Strathclyde, Glasgow, United Kingdom
| | - Andrea Slachevsky
- Gerosciences Center for Brain Health and Metabolism, Santiago, Chile
- Faculty of Medicine, University of Chile, Santiago, Chile
- Memory and Neuropsychiatric Clinic (CMYN) Neurology Department, Hospital del Salvador and University of Chile, Santiago, Chile
- Servicio de Neurología, Departamento de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago de Chile, Chile
| | - Gonzalo Farías
- Faculty of Medicine, University of Chile, Santiago, Chile
| | - Josefina Cruzat
- Latin American Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Adolfo García
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), CA, United States of America
- Cognitive Neuroscience Center (CNC), Universidad de San Andrés, Buenos Aires, Argentina
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- Departamento de Lingüística y Literatura, Facultad de Humanidades, Universidad de Santiago de Chile, Santiago, Chile
- Trinity College Dublin, Dublin, Ireland
| | - Harris A Eyre
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), CA, United States of America
- Neuroscience-Inspired Policy Initiative, Organisation for Economic Co-operation and Development and PRODEO Institute, Paris, France
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Victoria, Australia
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, United States of America
- Trinity College Dublin, Dublin, Ireland
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States of America
| | - Gil Rabinovici
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), CA, United States of America
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States of America
- Trinity College Dublin, Dublin, Ireland
| | - Robert Whelan
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), CA, United States of America
- Trinity College Dublin, Dublin, Ireland
| | - Agustín Ibáñez
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), CA, United States of America
- Cognitive Neuroscience Center (CNC), Universidad de San Andrés, Buenos Aires, Argentina
- Latin American Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
78
|
Chino-Vilca B, Concepción Rodríguez-Rojo I, Torres-Simón L, Cuesta P, Carnes Vendrell A, Piñol-Ripoll G, Huerto R, Tahan N, Maestú F. Sex specific EEG signatures associated with cerebrospinal fluid biomarkers in mild cognitive impairment. Clin Neurophysiol 2022; 142:190-198. [DOI: 10.1016/j.clinph.2022.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 06/07/2022] [Accepted: 08/06/2022] [Indexed: 11/25/2022]
|
79
|
Zhou J, Benoit M, Sharoar MG. Recent advances in pre-clinical diagnosis of Alzheimer's disease. Metab Brain Dis 2022; 37:1703-1725. [PMID: 33900524 DOI: 10.1007/s11011-021-00733-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 04/05/2021] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) is the most common dementia with currently no known cures or disease modifying treatments (DMTs), despite much time and effort from the field. Diagnosis and intervention of AD during the early pre-symptomatic phase of the disease is thought to be a more effective strategy. Therefore, the detection of biomarkers has emerged as a critical tool for monitoring the effect of new AD therapies, as well as identifying patients most likely to respond to treatment. The establishment of the amyloid/tau/neurodegeneration (A/T/N) framework in 2018 has codified the contexts of use of AD biomarkers in neuroimaging and bodily fluids for research and diagnostic purposes. Furthermore, a renewed drive for novel AD biomarkers and innovative methods of detection has emerged with the goals of adding additional insight to disease progression and discovery of new therapeutic targets. The use of biomarkers has accelerated the development of AD drugs and will bring new therapies to patients in need. This review highlights recent methods utilized to diagnose antemortem AD.
Collapse
Affiliation(s)
- John Zhou
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06030, USA
- Molecular Medicine Program, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Marc Benoit
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Md Golam Sharoar
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06030, USA.
| |
Collapse
|
80
|
Cersonsky TE, Mechery S, Carper MM, Thompson L, Lee A, Alber J, Sarkar IN, Brick LAD. Using the Montreal cognitive assessment to identify individuals with subtle cognitive decline. Neuropsychology 2022; 36:373-383. [PMID: 35511561 PMCID: PMC9912279 DOI: 10.1037/neu0000820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
OBJECTIVE Dementia is a devastating neurological disease that may be better managed if diagnosed earlier when subclinical neurodegenerative changes are already present, including subtle cognitive decline and mild cognitive impairment. In this study, we used item-level performance on the Montreal Cognitive Assessment (MoCA) to identify individuals with subtle cognitive decline. METHOD Individual MoCA item data from the Alzheimer's Disease Neuroimaging Initiative was grouped using k-modes cluster analysis. These clusters were validated and examined for association with convergent neuropsychological tests. The clusters were then compared and characterized using multinomial logistic regression. RESULTS A three-cluster solution had 77.3% precision, with Cluster 1 (high performing) displaying no deficits in performance, Cluster 2 (memory deficits) displaying lower memory performance, and Cluster 3 (compound deficits) displaying lower performance on memory and executive function. Age at MoCA (older in compound deficits), gender (more females in memory deficits), and marital status (fewer married in compound deficits) were significantly different among clusters. Age was not associated with increased odds of membership in the high-performing cluster compared to the others. CONCLUSIONS We identified three clusters of individuals classified as cognitively unimpaired using cluster analysis. Individuals in the compound deficits cluster performed lower on the MoCA and were older and less often married than individuals in other clusters. Demographic analyses suggest that cluster identity was due to a combination of both cognitive and clinical factors. Identifying individuals at risk for future cognitive decline using the MoCA could help them receive earlier evidence-based interventions to slow further cognitive decline. (PsycInfo Database Record (c) 2022 APA, all rights reserved).
Collapse
Affiliation(s)
- Tess E.K. Cersonsky
- Warren Alpert Medical School of Brown University, Providence, RI, USA
- Center for Biomedical Informatics, Brown University, Providence, RI, USA
| | - Shanti Mechery
- Memory and Aging Program, Butler Hospital, Providence, RI, USA
| | - Matthew M. Carper
- Warren Alpert Medical School of Brown University, Providence, RI, USA
- Quantitative Sciences Program, Department of Psychiatry and Neurology, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Emma Pendleton Bradley Hospital, Riverside, RI, USA
| | - Louisa Thompson
- Warren Alpert Medical School of Brown University, Providence, RI, USA
- Memory and Aging Program, Butler Hospital, Providence, RI, USA
| | - Athene Lee
- Warren Alpert Medical School of Brown University, Providence, RI, USA
- Memory and Aging Program, Butler Hospital, Providence, RI, USA
- Quantitative Sciences Program, Department of Psychiatry and Neurology, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Jessica Alber
- Memory and Aging Program, Butler Hospital, Providence, RI, USA
- Department of Biomedical and Pharmaceutical Sciences, George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| | - Indra Neil Sarkar
- Warren Alpert Medical School of Brown University, Providence, RI, USA
- Center for Biomedical Informatics, Brown University, Providence, RI, USA
- School of Public Health, Brown University, Providence, RI, USA
- Rhode Island Quality Institute, Providence, RI, USA
| | - Leslie Ann D. Brick
- Warren Alpert Medical School of Brown University, Providence, RI, USA
- Quantitative Sciences Program, Department of Psychiatry and Neurology, Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
81
|
Predictive Scale for Amyloid PET Positivity Based on Clinical and MRI Variables in Patients with Amnestic Mild Cognitive Impairment. J Clin Med 2022; 11:jcm11123433. [PMID: 35743503 PMCID: PMC9224873 DOI: 10.3390/jcm11123433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/11/2022] [Accepted: 06/12/2022] [Indexed: 12/05/2022] Open
Abstract
The presence of amyloid-β (Aβ) deposition is considered important in patients with amnestic mild cognitive impairment (aMCI), since they can progress to Alzheimer’s disease dementia. Amyloid positron emission tomography (PET) has been used for detecting Aβ deposition, but its high cost is a significant barrier for clinical usage. Therefore, we aimed to develop a new predictive scale for amyloid PET positivity using easily accessible tools. Overall, 161 aMCI patients were recruited from six memory clinics and underwent neuropsychological tests, brain magnetic resonance imaging (MRI), apolipoprotein E (APOE) genotype testing, and amyloid PET. Among the potential predictors, verbal and visual memory tests, medial temporal lobe atrophy, APOE genotype, and age showed significant differences between the Aβ-positive and Aβ-negative groups and were combined to make a model for predicting amyloid PET positivity with the area under the curve (AUC) of 0.856. Based on the best model, we developed the new predictive scale comprising integers, which had an optimal cutoff score ≥ 3. The new predictive scale was validated in another cohort of 98 participants and showed a good performance with AUC of 0.835. This new predictive scale with accessible variables may be useful for predicting Aβ positivity in aMCI patients in clinical practice.
Collapse
|
82
|
Laczó M, Martinkovic L, Lerch O, Wiener JM, Kalinova J, Matuskova V, Nedelska Z, Vyhnalek M, Hort J, Laczó J. Different Profiles of Spatial Navigation Deficits In Alzheimer’s Disease Biomarker-Positive Versus Biomarker-Negative Older Adults With Amnestic Mild Cognitive Impairment. Front Aging Neurosci 2022; 14:886778. [PMID: 35721017 PMCID: PMC9201637 DOI: 10.3389/fnagi.2022.886778] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/29/2022] [Indexed: 12/22/2022] Open
Abstract
BackgroundSpatial navigation impairment is a promising cognitive marker of Alzheimer’s disease (AD) that can reflect the underlying pathology.ObjectivesWe assessed spatial navigation performance in AD biomarker positive older adults with amnestic mild cognitive impairment (AD aMCI) vs. those AD biomarker negative (non-AD aMCI), and examined associations between navigation performance, MRI measures of brain atrophy, and cerebrospinal fluid (CSF) biomarkers.MethodsA total of 122 participants with AD aMCI (n = 33), non-AD aMCI (n = 31), mild AD dementia (n = 28), and 30 cognitively normal older adults (CN) underwent cognitive assessment, brain MRI (n = 100 had high-quality images for volumetric analysis) and three virtual navigation tasks focused on route learning (body-centered navigation), wayfinding (world-centered navigation) and perspective taking/wayfinding. Cognitively impaired participants underwent CSF biomarker assessment [amyloid-β1–42, total tau, and phosphorylated tau181 (p-tau181)] and amyloid PET imaging (n = 47 and n = 45, respectively), with a subset having both (n = 19).ResultsIn route learning, AD aMCI performed worse than non-AD aMCI (p < 0.001), who performed similarly to CN. In wayfinding, aMCI participants performed worse than CN (both p ≤ 0.009) and AD aMCI performed worse than non-AD aMCI in the second task session (p = 0.032). In perspective taking/wayfinding, aMCI participants performed worse than CN (both p ≤ 0.001). AD aMCI and non-AD aMCI did not differ in conventional cognitive tests. Route learning was associated with parietal thickness and amyloid-β1–42, wayfinding was associated with posterior medial temporal lobe (MTL) volume and p-tau181 and perspective taking/wayfinding was correlated with MRI measures of several brain regions and all CSF biomarkers.ConclusionAD biomarker positive and negative older adults with aMCI had different profiles of spatial navigation deficits that were associated with posterior MTL and parietal atrophy and reflected AD pathology.
Collapse
Affiliation(s)
- Martina Laczó
- Memory Clinic, Department of Neurology, Charles University, Second Faculty of Medicine and Motol University Hospital, Prague, Czechia
| | - Lukas Martinkovic
- Memory Clinic, Department of Neurology, Charles University, Second Faculty of Medicine and Motol University Hospital, Prague, Czechia
| | - Ondrej Lerch
- Memory Clinic, Department of Neurology, Charles University, Second Faculty of Medicine and Motol University Hospital, Prague, Czechia
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czechia
| | - Jan M. Wiener
- Department of Psychology, Ageing and Dementia Research Centre, Bournemouth University, Poole, United Kingdom
| | - Jana Kalinova
- Memory Clinic, Department of Neurology, Charles University, Second Faculty of Medicine and Motol University Hospital, Prague, Czechia
| | - Veronika Matuskova
- Memory Clinic, Department of Neurology, Charles University, Second Faculty of Medicine and Motol University Hospital, Prague, Czechia
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czechia
| | - Zuzana Nedelska
- Memory Clinic, Department of Neurology, Charles University, Second Faculty of Medicine and Motol University Hospital, Prague, Czechia
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czechia
| | - Martin Vyhnalek
- Memory Clinic, Department of Neurology, Charles University, Second Faculty of Medicine and Motol University Hospital, Prague, Czechia
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czechia
| | - Jakub Hort
- Memory Clinic, Department of Neurology, Charles University, Second Faculty of Medicine and Motol University Hospital, Prague, Czechia
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czechia
| | - Jan Laczó
- Memory Clinic, Department of Neurology, Charles University, Second Faculty of Medicine and Motol University Hospital, Prague, Czechia
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czechia
- *Correspondence: Jan Laczó
| |
Collapse
|
83
|
Amft M, Ortner M, Eichenlaub U, Goldhardt O, Diehl-Schmid J, Hedderich DM, Yakushev I, Grimmer T. The cerebrospinal fluid biomarker ratio Aβ42/40 identifies amyloid positron emission tomography positivity better than Aβ42 alone in a heterogeneous memory clinic cohort. Alzheimers Res Ther 2022; 14:60. [PMID: 35473631 PMCID: PMC9044878 DOI: 10.1186/s13195-022-01003-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 04/08/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Cerebrospinal fluid (CSF) analysis for detecting amyloid positivity may be as reliable as positron emission tomography (PET). We evaluated the performance of the amyloid beta (Aβ)42/40 ratio for predicting amyloid positivity by PET, compared with Aβ42 alone, and phosphorylated tau 181 (pTau181)/Aβ42 and total tau (tTau)/Aβ42 ratios, using fully automated CSF immunoassays (Roche Diagnostics International Ltd, Rotkreuz, Switzerland) in a heterogeneous cohort of patients with a range of cognitive disorders reflecting the typical population of a memory clinic. METHODS CSF samples from 103 patients with known amyloid PET status (PET positive = 54; PET negative = 49) were retrospectively selected from one site in Germany; 71 patients were undergoing treatment for mild cognitive impairment (n = 44) or mild-to-moderate dementia (n = 27) due to Alzheimer's disease (AD), and 32 patients were undergoing treatment for non-AD-related cognitive disorders. Aβ42, pTau181, and tTau concentrations were measured in CSF samples using the respective Elecsys® CSF immunoassays modified for use on the cobas e 411 analyzer; Aβ40 concentrations were measured using a non-commercially available robust prototype assay. Sensitivities/specificities for amyloid positivity cut-offs (Youden-derived and pre-defined) were calculated, and receiver operating characteristic analyses determined area under the curve (AUC) versus amyloid PET status. Limitations include a small sample size, use of a pre-analytical protocol not in accordance with the Elecsys CSF immunoassay method sheets, and the lack of a pre-defined cut-off for Aβ42/40. RESULTS Point estimates for sensitivity and specificity of CSF biomarkers and biomarker ratios versus amyloid PET were 0.93 and 0.57 for Aβ42, 0.96 and 0.69 for pTau181/Aβ42, 0.92 and 0.69 for tTau/Aβ42, and 0.94 and 0.82 for Aβ42/40. For AUCs, point estimates (95% confidence intervals) versus amyloid PET were 0.78 (0.68-0.88) for Aβ42, 0.88 (0.81-0.95) for pTau181/Aβ42, 0.87 (0.80-0.95) for tTau/Aβ42, and 0.90 (0.83-0.97) for Aβ42/40. CONCLUSIONS CSF Aβ42/40 ratio can predict PET amyloid positivity with high accuracy in patients with a range of cognitive disorders when evaluating Aβ pathology independent of tau and neurodegeneration for research purposes. The performance of Aβ42/40 was comparable with pTau181/Aβ42 and tTau/Aβ42 used in clinical practice and better than Aβ42 alone.
Collapse
Affiliation(s)
- Michaela Amft
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Marion Ortner
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Udo Eichenlaub
- Clinical Development Medical Affairs, Roche Diagnostic Solutions, Roche Diagnostics GmbH, Penzberg, Germany
| | - Oliver Goldhardt
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Janine Diehl-Schmid
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Dennis M Hedderich
- Department of Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Igor Yakushev
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Timo Grimmer
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany.
| |
Collapse
|
84
|
Klyucherev TO, Olszewski P, Shalimova AA, Chubarev VN, Tarasov VV, Attwood MM, Syvänen S, Schiöth HB. Advances in the development of new biomarkers for Alzheimer's disease. Transl Neurodegener 2022; 11:25. [PMID: 35449079 PMCID: PMC9027827 DOI: 10.1186/s40035-022-00296-z] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 03/28/2022] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is a complex, heterogeneous, progressive disease and is the most common type of neurodegenerative dementia. The prevalence of AD is expected to increase as the population ages, placing an additional burden on national healthcare systems. There is a large need for new diagnostic tests that can detect AD at an early stage with high specificity at relatively low cost. The development of modern analytical diagnostic tools has made it possible to determine several biomarkers of AD with high specificity, including pathogenic proteins, markers of synaptic dysfunction, and markers of inflammation in the blood. There is a considerable potential in using microRNA (miRNA) as markers of AD, and diagnostic studies based on miRNA panels suggest that AD could potentially be determined with high accuracy for individual patients. Studies of the retina with improved methods of visualization of the fundus are also showing promising results for the potential diagnosis of the disease. This review focuses on the recent developments of blood, plasma, and ocular biomarkers for the diagnosis of AD.
Collapse
Affiliation(s)
- Timofey O Klyucherev
- Department of Neuroscience, Functional Pharmacology, University of Uppsala, Uppsala, Sweden.,Department of Pharmacology, Institute of Pharmacy, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Pawel Olszewski
- Department of Neuroscience, Functional Pharmacology, University of Uppsala, Uppsala, Sweden
| | - Alena A Shalimova
- Department of Neuroscience, Functional Pharmacology, University of Uppsala, Uppsala, Sweden.,Department of Pharmacology, Institute of Pharmacy, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vladimir N Chubarev
- Institute of Translational Medicine and Biotechnology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vadim V Tarasov
- Department of Pharmacology, Institute of Pharmacy, I. M. Sechenov First Moscow State Medical University, Moscow, Russia.,Institute of Translational Medicine and Biotechnology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Misty M Attwood
- Department of Neuroscience, Functional Pharmacology, University of Uppsala, Uppsala, Sweden
| | - Stina Syvänen
- Department of Public Health and Caring Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Helgi B Schiöth
- Department of Neuroscience, Functional Pharmacology, University of Uppsala, Uppsala, Sweden.
| |
Collapse
|
85
|
Day GS, Scarmeas N, Dubinsky R, Coerver K, Mostacero A, West B, Wessels SR, Armstrong MJ. Aducanumab Use in Symptomatic Alzheimer Disease Evidence in Focus: A Report of the AAN Guidelines Subcommittee. Neurology 2022; 98:619-631. [PMID: 35197360 PMCID: PMC9012273 DOI: 10.1212/wnl.0000000000200176] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/25/2022] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To identify the class of evidence for aducanumab use for the treatment of Alzheimer disease and present clinical considerations regarding use. METHODS The author panel systematically reviewed available clinical trial data detailing aducanumab use in individuals with early symptomatic Alzheimer disease. Level of evidence statements were assigned in accordance with the American Academy of Neurology's 2017 therapeutic classification of evidence scheme. Safety information, regulatory decisions, and clinical context were also reviewed. RESULTS Data were identified from 4 clinical trials, 1 rated Class I and 3 rated Class II. The Class I study showed that single doses of aducanumab up to 30 mg/kg were safe and well tolerated. All 3 Class II studies provided evidence that aducanumab (3-10 mg/kg) decreased amyloid deposition on brain PET at 1 year vs placebo. Efficacy data in the Class II studies varied by dose and outcome, but aducanumab either had no effect on mean change on the Clinical Dementia Rating Sum of Boxes scores or resulted in less worsening (vs placebo) that was of uncertain clinical importance. Adverse amyloid-related imaging abnormalities occurred in approximately 40% of individuals treated with aducanumab vs 10% receiving placebo. CLINICAL CONTEXT Administration of aducanumab will require expanded clinical infrastructure. Evidence-based guidance is needed to address key questions (e.g., safety in populations not enrolled in phase 3 studies, expected benefits on daily function, treatment duration) and critical issues relating to access to aducanumab (e.g., coverage, costs, burden of monthly infusions) that will inform shared decision making between patients and providers.
Collapse
Affiliation(s)
- Gregory S Day
- From the Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL; National and Kapodistrian University of Athens (N.S.), Greece; Columbia University (N.S.), New York, NY; University of Kansas Medical Center (R.D.), Kansas City; Rocky Mountain Neurology (K.C.), Lone Tree, CO; Patient Representative (A.M.), San Antonio, TX; Care Partner Representative (B.W.), Telluride, CO; American Academy of Neurology (S.R.W.), Minneapolis, MN; and Department of Neurology (M.J.A.), University of Florida, Gainesville
| | - Nikolaos Scarmeas
- From the Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL; National and Kapodistrian University of Athens (N.S.), Greece; Columbia University (N.S.), New York, NY; University of Kansas Medical Center (R.D.), Kansas City; Rocky Mountain Neurology (K.C.), Lone Tree, CO; Patient Representative (A.M.), San Antonio, TX; Care Partner Representative (B.W.), Telluride, CO; American Academy of Neurology (S.R.W.), Minneapolis, MN; and Department of Neurology (M.J.A.), University of Florida, Gainesville
| | - Richard Dubinsky
- From the Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL; National and Kapodistrian University of Athens (N.S.), Greece; Columbia University (N.S.), New York, NY; University of Kansas Medical Center (R.D.), Kansas City; Rocky Mountain Neurology (K.C.), Lone Tree, CO; Patient Representative (A.M.), San Antonio, TX; Care Partner Representative (B.W.), Telluride, CO; American Academy of Neurology (S.R.W.), Minneapolis, MN; and Department of Neurology (M.J.A.), University of Florida, Gainesville
| | - Katherine Coerver
- From the Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL; National and Kapodistrian University of Athens (N.S.), Greece; Columbia University (N.S.), New York, NY; University of Kansas Medical Center (R.D.), Kansas City; Rocky Mountain Neurology (K.C.), Lone Tree, CO; Patient Representative (A.M.), San Antonio, TX; Care Partner Representative (B.W.), Telluride, CO; American Academy of Neurology (S.R.W.), Minneapolis, MN; and Department of Neurology (M.J.A.), University of Florida, Gainesville
| | - Anitra Mostacero
- From the Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL; National and Kapodistrian University of Athens (N.S.), Greece; Columbia University (N.S.), New York, NY; University of Kansas Medical Center (R.D.), Kansas City; Rocky Mountain Neurology (K.C.), Lone Tree, CO; Patient Representative (A.M.), San Antonio, TX; Care Partner Representative (B.W.), Telluride, CO; American Academy of Neurology (S.R.W.), Minneapolis, MN; and Department of Neurology (M.J.A.), University of Florida, Gainesville
| | - Brooks West
- From the Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL; National and Kapodistrian University of Athens (N.S.), Greece; Columbia University (N.S.), New York, NY; University of Kansas Medical Center (R.D.), Kansas City; Rocky Mountain Neurology (K.C.), Lone Tree, CO; Patient Representative (A.M.), San Antonio, TX; Care Partner Representative (B.W.), Telluride, CO; American Academy of Neurology (S.R.W.), Minneapolis, MN; and Department of Neurology (M.J.A.), University of Florida, Gainesville
| | - Scott R Wessels
- From the Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL; National and Kapodistrian University of Athens (N.S.), Greece; Columbia University (N.S.), New York, NY; University of Kansas Medical Center (R.D.), Kansas City; Rocky Mountain Neurology (K.C.), Lone Tree, CO; Patient Representative (A.M.), San Antonio, TX; Care Partner Representative (B.W.), Telluride, CO; American Academy of Neurology (S.R.W.), Minneapolis, MN; and Department of Neurology (M.J.A.), University of Florida, Gainesville
| | - Melissa J Armstrong
- From the Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL; National and Kapodistrian University of Athens (N.S.), Greece; Columbia University (N.S.), New York, NY; University of Kansas Medical Center (R.D.), Kansas City; Rocky Mountain Neurology (K.C.), Lone Tree, CO; Patient Representative (A.M.), San Antonio, TX; Care Partner Representative (B.W.), Telluride, CO; American Academy of Neurology (S.R.W.), Minneapolis, MN; and Department of Neurology (M.J.A.), University of Florida, Gainesville
| |
Collapse
|
86
|
Giorgio J, Jagust WJ, Baker S, Landau SM, Tino P, Kourtzi Z. A robust and interpretable machine learning approach using multimodal biological data to predict future pathological tau accumulation. Nat Commun 2022; 13:1887. [PMID: 35393421 PMCID: PMC8989879 DOI: 10.1038/s41467-022-28795-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 02/11/2022] [Indexed: 01/21/2023] Open
Abstract
The early stages of Alzheimer's disease (AD) involve interactions between multiple pathophysiological processes. Although these processes are well studied, we still lack robust tools to predict individualised trajectories of disease progression. Here, we employ a robust and interpretable machine learning approach to combine multimodal biological data and predict future pathological tau accumulation. In particular, we use machine learning to quantify interactions between key pathological markers (β-amyloid, medial temporal lobe atrophy, tau and APOE 4) at mildly impaired and asymptomatic stages of AD. Using baseline non-tau markers we derive a prognostic index that: (a) stratifies patients based on future pathological tau accumulation, (b) predicts individualised regional future rate of tau accumulation, and (c) translates predictions from deep phenotyping patient cohorts to cognitively normal individuals. Our results propose a robust approach for fine scale stratification and prognostication with translation impact for clinical trial design targeting the earliest stages of AD.
Collapse
Affiliation(s)
- Joseph Giorgio
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
- Molecular Biophysics & Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Suzanne Baker
- Molecular Biophysics & Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Susan M Landau
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Peter Tino
- School of Computer Science, University of Birmingham, Birmingham, UK
| | - Zoe Kourtzi
- Department of Psychology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
87
|
Uchida K. Waste Clearance in the Brain and Neuroinflammation: A Novel Perspective on Biomarker and Drug Target Discovery in Alzheimer's Disease. Cells 2022; 11:cells11050919. [PMID: 35269541 PMCID: PMC8909773 DOI: 10.3390/cells11050919] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/26/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is a multifactorial disease with a heterogeneous etiology. The pathology of Alzheimer’s disease is characterized by amyloid-beta and hyperphosphorylated tau, which are necessary for disease progression. Many clinical trials on disease-modifying drugs for AD have failed to indicate their clinical benefits. Recent advances in fundamental research have indicated that neuroinflammation plays an important pathological role in AD. Damage- and pathogen-associated molecular patterns in the brain induce neuroinflammation and inflammasome activation, causing caspase-1-dependent glial and neuronal cell death. These waste products in the brain are eliminated by the glymphatic system via perivascular spaces, the blood-brain barrier, and the blood–cerebrospinal fluid barrier. Age-related vascular dysfunction is associated with an impairment of clearance and barrier functions, leading to neuroinflammation. The proteins involved in waste clearance in the brain and peripheral circulation may be potential biomarkers and drug targets in the early stages of cognitive impairment. This short review focuses on waste clearance dysfunction in AD pathobiology and discusses the improvement of waste clearance as an early intervention in prodromal AD and preclinical stages of dementia.
Collapse
Affiliation(s)
- Kazuhiko Uchida
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba 305-8575, Ibaraki, Japan; ; Tel.: +81-29-853-3210; Fax: +81-50-3730-7456
- Institute for Biomedical Research, MCBI, 4-9-29 Matsushiro, Tsukuba 305-0035, Ibaraki, Japan
| |
Collapse
|
88
|
Woodfield A, Porter T, Gilani I, Noordin S, Li QX, Collins S, Martins RN, Maruff P, Masters CL, Rowe CC, Villemagne VL, Dore V, Newsholme P, Laws SM, Verdile G. Insulin resistance, cognition and Alzheimer's disease biomarkers: Evidence that CSF Aβ42 moderates the association between insulin resistance and increased CSF tau levels. Neurobiol Aging 2022; 114:38-48. [DOI: 10.1016/j.neurobiolaging.2022.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/09/2022] [Accepted: 03/07/2022] [Indexed: 12/16/2022]
|
89
|
Lazar SV, Mor S, Wang D, Goldbloom-Helzner L, Clark K, Hao D, Farmer DL, Wang A. Engineering extracellular vesicles for Alzheimer's disease: An emerging cell-free approach for earlier diagnosis and treatment. WIREs Mech Dis 2022; 14:e1541. [PMID: 35266650 PMCID: PMC9397584 DOI: 10.1002/wsbm.1541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/20/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative disorder affecting over five million people globally and has no established cure. Current AD-related treatments only alleviate cognitive and behavioral symptoms and do not address disease onset or progression, underlining the unmet need to create an effective, innovative AD therapeutic. Extracellular vesicles (EVs) have emerged as a new class of nanotherapeutics. These secreted, lipid-bound cellular signaling carriers show promise for potential clinical applications for neurodegenerative diseases like AD. Additionally, analyzing contents and characteristics of patient-derived EVs may address the unmet need for earlier AD diagnostic techniques, informing physicians of altered genetic expression or cellular communications specific to healthy and diseased physiological states. There are numerous recent advances in regenerative medicine using EVs and include bioengineering perspectives to modify EVs, target glial cells in neurodegenerative diseases like AD, and potentially use EVs to diagnose and treat AD earlier. This article is categorized under: Neurological Diseases > Biomedical Engineering Neurological Diseases > Molecular and Cellular Physiology Neurological Diseases > Stem Cells and Development.
Collapse
Affiliation(s)
| | - Sirjan Mor
- Department of Surgery, University of California, Davis
| | - David Wang
- Department of Surgery, Department of Biomedical Engineering, University of California, Davis
| | - Leora Goldbloom-Helzner
- Department of Surgery, Department of Biomedical Engineering, University of California, Davis
| | - Kaitlin Clark
- Department of Surgery, University of California, Davis
| | - Dake Hao
- Department of Surgery, Shriners Hospitals for Children Northern California – Institute for Pediatric Regenerative Medicine, University of California, Davis
| | - Diana Lee Farmer
- Department of Surgery, Shriners Hospitals for Children Northern California – Institute for Pediatric Regenerative Medicine, University of California, Davis
| | - Aijun Wang
- Department of Surgery, Department of Biomedical Engineering, Shriners Hospitals for Children Northern California – Institute for Pediatric Regenerative Medicine, University of California, Davis
| |
Collapse
|
90
|
Evidence of plasma biomarkers indicating high risk of dementia in cognitively normal subjects. Sci Rep 2022; 12:1192. [PMID: 35075194 PMCID: PMC8786959 DOI: 10.1038/s41598-022-05177-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 01/07/2022] [Indexed: 11/08/2022] Open
Abstract
Subjects with comorbidities are at risk for neurodegeneration. There is a lack of a direct relationship between comorbidities and neurodegeneration. In this study, immunomagnetic reduction (IMR) assays were utilized to assay plasma Aβ1-42 and total tau protein (T-Tau) levels in poststroke (PS, n = 27), family history of Alzheimer's disease (ADFH, n = 35), diabetes (n = 21), end-stage renal disease (ESRD, n = 41), obstructive sleep apnea (OSA, n = 20), Alzheimer's disease (AD, n = 65). Thirty-seven healthy controls (HCs) were enrolled. The measured concentrations of plasma Aβ1-42 were 14.26 ± 1.42, 15.43 ± 1.76, 15.52 ± 1.60, 16.15 ± 1.05, 16.52 ± 0.59, 15.97 ± 0.54 and 20.06 ± 3.09 pg/mL in HC, PS, ADFH, diabetes, ESRD, OSA and AD groups, respectively. The corresponding concentrations of plasma T-Tau were 15.13 ± 3.62, 19.29 ± 8.01, 17.93 ± 6.26, 19.74 ± 2.92, 21.54 ± 2.72, 20.17 ± 2.77 and 41.24 ± 14.64 pg/mL. The plasma levels of Aβ1-42 and T-Tau in were significantly higher in the PS, ADFH, diabetes, ESRD and OSA groups than controls (Aβ1-42 in PS: 15.43 ± 1.76 pg/mL vs. 14.26 ± 1.42 pg/mL, p < 0.005; T-Tau in PS: 19.29 ± 8.01 vs. 15.13 ± 3.62 pg/mL, p < 0.005, Aβ1-42 in ADFH: 15.52 ± 1.60 pg/mL vs. 14.26 ± 1.42 pg/mL, p < 0.001; T-Tau in ADFH: 17.93 ± 6.26 vs. 15.13 ± 3.62 pg/mL, p < 0.005, Aβ1-42 in diabetes: 16.15 ± 1.05 pg/mL vs. 14.26 ± 1.42 pg/mL, p < 0.001; T-Tau in diabetes: 19.74 ± 2.92 vs. 15.13 ± 3.62 pg/mL, p < 0.001, Aβ1-42 in ESRD: 16.52 ± 0.59 pg/mL vs. 14.26 ± 1.42 pg/mL, p < 0.001; T-Tau in ESRD: 21.54 ± 2.72 vs. 15.13 ± 3.62 pg/mL, p < 0.001, Aβ1-42 in OSA: 15.97 ± 0.54 pg/mL vs. 14.26 ± 1.42 pg/mL, p < 0.001; T-Tau in OSA: 20.17 ± 2.77 vs. 15.13 ± 3.62 pg/mL, p < 0.001). This evidence indicates the high risk for dementia in these groups from the perspective of plasma biomarkers.
Collapse
|
91
|
Cummings J, Rabinovici GD, Atri A, Aisen P, Apostolova LG, Hendrix S, Sabbagh M, Selkoe D, Weiner M, Salloway S. Aducanumab: Appropriate Use Recommendations Update. J Prev Alzheimers Dis 2022; 9:221-230. [PMID: 35542993 PMCID: PMC9169517 DOI: 10.14283/jpad.2022.34] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Aducanumab (Aduhelm) is approved in the United States for the treatment of patients with mild cognitive impairment due to Alzheimer's disease or mild AD dementia. Aducanumab Appropriate Use Recommendations (AURs) have been published and have helped guide best practices for use of aducanumab. As real-world use has occurred and more information has accrued, the AURs require refinement. We update the AURs to better inform appropriate patient selection and improve shared decision-making, safety monitoring, and risk mitigation in treated patients. Based on evolving experience we emphasize the importance of detecting past medical conditions that may predispose to amyloid related imaging abnormalities (ARIA) or may increase the likelihood of ARIA complications including autoimmune or inflammatory conditions, seizures, or disorders associated with extensive white matter pathology. The apolipoprotein E ε4 (APOE4) genotype is strongly associated with ARIA and exhibits a gene dose effect. We recommend that clinicians perform APOE genotyping to better inform patient care decisions, discussions regarding risk, and clinician vigilance concerning ARIA. As most ARIA occurs during the titration period of aducanumab, we suggest performing MRI before the 5th, 7th, 9th, and 12th infusions to improve detection. Uncommonly, ARIA may be recurrent or serious; we suggest additional parameters for treatment discontinuation taking these observations into account. It is important to continue to learn from the real-world use of aducanumab and the AURs will continue to evolve as new information becomes available. This AUR update does not address efficacy, price, or insurance coverage and is provided to assist clinicians to establish best practices for use of aducanumab in the treatment of patients with mild cognitive impairment and mild Alzheimer's dementia.
Collapse
Affiliation(s)
- J Cummings
- Jeffrey Cummings, MD, ScD, 1380 Opal Valley Street, Henderson, NV 89052, USA, , T: 702-902-3939
| | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Pavlik VN, Burnham SC, Kass JS, Helmer C, Palmqvist S, Vassilaki M, Dartigues JF, Hansson O, Masters CL, Pérès K, Petersen RC, Stomrud E, Butler L, Coloma PM, Teitsma XM, Doody R, Sano M. Connecting Cohorts to Diminish Alzheimer's Disease (CONCORD-AD): A Report of an International Research Collaboration Network. J Alzheimers Dis 2022; 85:31-45. [PMID: 34776434 PMCID: PMC8842789 DOI: 10.3233/jad-210525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2021] [Indexed: 11/15/2022]
Abstract
Longitudinal observational cohort studies are being conducted worldwide to understand cognition, biomarkers, and the health of the aging population better. Cross-cohort comparisons and networks of registries in Alzheimer's disease (AD) foster scientific exchange, generate insights, and contribute to the evolving clinical science in AD. A scientific working group was convened with invited investigators from established cohort studies in AD, in order to form a research collaboration network as a resource to address important research questions. The Connecting Cohorts to Diminish Alzheimer's Disease (CONCORD-AD) collaboration network was created to bring together global resources and expertise, to generate insights and improve understanding of the natural history of AD, to inform design of clinical trials in all disease stages, and to plan for optimal patient access to disease-modifying therapies once they become available. The network brings together expertise and data insights from 7 cohorts across Australia, Europe, and North America. Notably, the network includes populations recruited through memory clinics as well as population-based cohorts, representing observations from individuals across the AD spectrum. This report aims to introduce the CONCORD-AD network, providing an overview of the cohorts involved, reporting the common assessments used, and describing the key characteristics of the cohort populations. Cohort study designs and baseline population characteristics are compared, and available cognitive, functional, and neuropsychiatric symptom data, as well as the frequency of biomarker assessments, are summarized. Finally, the challenges and opportunities of cross-cohort studies in AD are discussed.
Collapse
Affiliation(s)
- Valory N. Pavlik
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Samantha C. Burnham
- The Australian eHealth Research Centre, CSIRO Health and Biosecurity, Melbourne, VIC, Australia
| | - Joseph S. Kass
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Catherine Helmer
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR, Bordeaux, France
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Maria Vassilaki
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - Jean-François Dartigues
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR, Bordeaux, France
- Department of Neurology, Memory Consultation, Bordeaux University Hospital, Bordeaux, France
| | - Oskar Hansson
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Colin L. Masters
- The Florey Institute and The University of Melbourne, Parkville, VIC, Australia
| | - Karine Pérès
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR, Bordeaux, France
| | - Ronald C. Petersen
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Erik Stomrud
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Lesley Butler
- Product Development Personalised Health Care – Data Science, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Preciosa M. Coloma
- Product Development Personalised Health Care – Data Science, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Xavier M. Teitsma
- Product Development Personalised Health Care – Data Science, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Rachelle Doody
- Product Development Neuroscience, F. Hoffmann-La Roche Ltd, Basel, Switzerland
- Product Development Neuroscience, Genentech, Inc., South San Francisco, CA, USA
| | - Mary Sano
- Department of Psychiatry, Alzheimer’s Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - for the CONCORD-AD investigators
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
- The Australian eHealth Research Centre, CSIRO Health and Biosecurity, Melbourne, VIC, Australia
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR, Bordeaux, France
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Memory Consultation, Bordeaux University Hospital, Bordeaux, France
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
- The Florey Institute and The University of Melbourne, Parkville, VIC, Australia
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Product Development Personalised Health Care – Data Science, F. Hoffmann-La Roche Ltd, Basel, Switzerland
- Product Development Neuroscience, F. Hoffmann-La Roche Ltd, Basel, Switzerland
- Product Development Neuroscience, Genentech, Inc., South San Francisco, CA, USA
- Department of Psychiatry, Alzheimer’s Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| |
Collapse
|
93
|
Sacchi L, Carandini T, Fumagalli GG, Pietroboni AM, Contarino VE, Siggillino S, Arcaro M, Fenoglio C, Zito F, Marotta G, Castellani M, Triulzi F, Galimberti D, Scarpini E, Arighi A. Unravelling the Association Between Amyloid-PET and Cerebrospinal Fluid Biomarkers in the Alzheimer's Disease Spectrum: Who Really Deserves an A+? J Alzheimers Dis 2021; 85:1009-1020. [PMID: 34897084 DOI: 10.3233/jad-210593] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Association between cerebrospinal fluid (CSF)-amyloid-β (Aβ)42 and amyloid-PET measures is inconstant across the Alzheimer's disease (AD) spectrum. However, they are considered interchangeable, along with Aβ 42/40 ratio, for defining 'Alzheimer's Disease pathologic change' (A+). OBJECTIVE Herein, we further characterized the association between amyloid-PET and CSF biomarkers and tested their agreement in a cohort of AD spectrum patients. METHODS We include ed 23 patients who underwent amyloid-PET, MRI, and CSF analysis showing reduced levels of Aβ 42 within a 365-days interval. Thresholds used for dichotomization were: Aβ 42 < 640 pg/mL (Aβ 42+); pTau > 61 pg/mL (pTau+); and Aβ 42/40 < 0.069 (ADratio+). Amyloid-PET scans were visually assessed and processed by four pipelines (SPMCL, SPMAAL, FSGM, FSWC). RESULTS Different pipelines gave highly inter-correlated standardized uptake value ratios (SUVRs) (rho = 0.93-0.99). The most significant findings were: pTau positive correlation with SPMCL SUVR (rho = 0.56, p = 0.0063) and Aβ 42/40 negative correlation with SPMCL and SPMAAL SUVRs (rho = -0.56, p = 0.0058; rho = -0.52, p = 0.0117 respectively). No correlations between CSF-Aβ 42 and global SUVRs were observed. In subregion analysis, both pTau and Aβ 42/40 values significantly correlated with cingulate SUVRs from any pipeline (R2 = 0.55-0.59, p < 0.0083), with the strongest associations observed for the posterior/isthmus cingulate areas. However, only associations observed for Aβ 42/40 ratio were still significant in linear regression models. Moreover, combining pTau with Aβ 42 or using Aβ 42/40, instead of Aβ 42 alone, increased concordance with amyloid-PET status from 74% to 91% based on visual reads and from 78% to 96% based on Centiloids. CONCLUSION We confirmed that, in the AD spectrum, amyloid-PET measures show a stronger association and a better agreement with CSF-Aβ 42/40 and secondarily pTau rather than Aβ 42 levels.
Collapse
Affiliation(s)
- Luca Sacchi
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Tiziana Carandini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Anna Margherita Pietroboni
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Silvia Siggillino
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marina Arcaro
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Fenoglio
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Felicia Zito
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giorgio Marotta
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Massimo Castellani
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Fabio Triulzi
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Galimberti
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elio Scarpini
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea Arighi
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
94
|
Andersen E, Casteigne B, Chapman WD, Creed A, Foster F, Lapins A, Shatz R, Sawyer RP. Diagnostic biomarkers in Alzheimer’s disease. Biomark Neuropsychiatry 2021. [DOI: 10.1016/j.bionps.2021.100041] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
95
|
Ricci M, Cimini A, Camedda R, Chiaravalloti A, Schillaci O. Tau Biomarkers in Dementia: Positron Emission Tomography Radiopharmaceuticals in Tauopathy Assessment and Future Perspective. Int J Mol Sci 2021; 22:ijms222313002. [PMID: 34884804 PMCID: PMC8657996 DOI: 10.3390/ijms222313002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/14/2021] [Accepted: 11/25/2021] [Indexed: 01/20/2023] Open
Abstract
Abnormal accumulation of Tau protein is closely associated with neurodegeneration and cognitive impairment and it is a biomarker of neurodegeneration in the dementia field, especially in Alzheimer’s disease (AD); therefore, it is crucial to be able to assess the Tau deposits in vivo. Beyond the fluid biomarkers of tauopathy described in this review in relationship with the brain glucose metabolic patterns, this review aims to focus on tauopathy assessment by using Tau PET imaging. In recent years, several first-generation Tau PET tracers have been developed and applied in the dementia field. Common limitations of first-generation tracers include off-target binding and subcortical white-matter uptake; therefore, several institutions are working on developing second-generation Tau tracers. The increasing knowledge about the distribution of first- and second-generation Tau PET tracers in the brain may support physicians with Tau PET data interpretation, both in the research and in the clinical field, but an updated description of differences in distribution patterns among different Tau tracers, and in different clinical conditions, has not been reported yet. We provide an overview of first- and second-generation tracers used in ongoing clinical trials, also describing the differences and the properties of novel tracers, with a special focus on the distribution patterns of different Tau tracers. We also describe the distribution patterns of Tau tracers in AD, in atypical AD, and further neurodegenerative diseases in the dementia field.
Collapse
Affiliation(s)
- Maria Ricci
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (A.C.); (R.C.); (A.C.); (O.S.)
- Correspondence:
| | - Andrea Cimini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (A.C.); (R.C.); (A.C.); (O.S.)
| | - Riccardo Camedda
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (A.C.); (R.C.); (A.C.); (O.S.)
| | - Agostino Chiaravalloti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (A.C.); (R.C.); (A.C.); (O.S.)
- Nuclear Medicine Section, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (A.C.); (R.C.); (A.C.); (O.S.)
- Nuclear Medicine Section, IRCCS Neuromed, 86077 Pozzilli, Italy
| |
Collapse
|
96
|
Yun G, Kim HJ, Kim HG, Lee KM, Hong IK, Kim SH, Rhee HY, Jahng GH, Yoon SS, Park KC, Hwang KS, Lee JS. Association Between Plasma Amyloid-β and Neuropsychological Performance in Patients With Cognitive Decline. Front Aging Neurosci 2021; 13:736937. [PMID: 34759814 PMCID: PMC8573146 DOI: 10.3389/fnagi.2021.736937] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/07/2021] [Indexed: 01/10/2023] Open
Abstract
Objective: To investigate the association between plasma amyloid-β (Aβ) levels and neuropsychological performance in patients with cognitive decline using a highly sensitive nano-biosensing platform. Methods: We prospectively recruited 44 patients with cognitive decline who underwent plasma Aβ analysis, amyloid positron emission tomography (PET) scanning, and detailed neuropsychological tests. Patients were classified into a normal control (NC, n = 25) or Alzheimer’s disease (AD, n = 19) group based on amyloid PET positivity. Multiple linear regression was performed to determine whether plasma Aβ (Aβ40, Aβ42, and Aβ42/40) levels were associated with neuropsychological test results. Results: The plasma levels of Aβ42/40 were significantly different between the NC and AD groups and were the best predictor of amyloid PET positivity by receiver operating characteristic curve analysis [area under the curve of 0.952 (95% confidence interval, 0.892–1.000)]. Although there were significant differences in the neuropsychological performance of cognitive domains (language, visuospatial, verbal/visual memory, and frontal/executive functions) between the NC and AD groups, higher levels of plasma Aβ42/40 were negatively correlated only with verbal and visual memory performance. Conclusion: Our results demonstrated that plasma Aβ analysis using a nano-biosensing platform could be a useful tool for diagnosing AD and assessing memory performance in patients with cognitive decline.
Collapse
Affiliation(s)
- Gyihyaon Yun
- Department of Neurology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Hye Jin Kim
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Hyug-Gi Kim
- Department of Radiology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Kyung Mi Lee
- Department of Radiology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Il Ki Hong
- Department of Nuclear Medicine, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Sang Hoon Kim
- Department of Otorhinolaryngology, Head and Neck Surgery, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Hak Young Rhee
- Department of Neurology, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Geon-Ho Jahng
- Department of Radiology, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Sung Sang Yoon
- Department of Neurology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Key-Chung Park
- Department of Neurology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Kyo Seon Hwang
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Jin San Lee
- Department of Neurology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| |
Collapse
|
97
|
Nerattini M, Rubino F, Arnone A, Polito C, Mazzeo S, Lombardi G, Puccini G, Nacmias B, De Cristofaro MT, Sorbi S, Pupi A, Sciagrà R, Bessi V, Berti V. Cerebral amyloid load determination in a clinical setting: interpretation of amyloid biomarker discordances aided by tau and neurodegeneration measurements. Neurol Sci 2021; 43:2469-2480. [PMID: 34739618 DOI: 10.1007/s10072-021-05704-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 10/26/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) diagnosis can be hindered by amyloid biomarkers discordances. OBJECTIVE We aim to interpret discordances between amyloid positron emission tomography (Amy-PET) and cerebrospinal fluid (CSF) (Aβ42 and Aβ42/40), using Amy-PET semiquantitative analysis, [18F]fluorodeoxyglucose (FDG)-PET pattern, and CSF assays. METHOD Thirty-six subjects with dementia or mild cognitive impairment, assessed by neuropsychological tests, structural and functional imaging, and CSF assays (Aβ42, Aβ42/40, p-tau, t-tau), were retrospectively examined. Amy-PET and FDG-PET scans were analyzed by visual assessment and voxel-based analysis. SUVR were calculated on Amy-PET scans. RESULTS Groups were defined basing on the agreement among CSF Aβ42 (A), CSF Aβ42/40 Ratio (R), and Amy-PET (P) dichotomic results ( ±). In discordant groups, CSF assays, Amy-PET semiquantification, and FDG-PET patterns supported the diagnosis suggested by any two agreeing amyloid biomarkers. In groups with discordant CSF Aβ42, the ratio always agrees with Amy-PET results, solving both false-negative and false-positive Aβ42 results, with Aβ42 levels close to the cut-off in A + R-P- subjects. The A + R + P- group presented high amyloid deposition in relevant areas, such as precuneus, posterior cingulate cortex (PCC) and dorsolateral frontal inferior cortex at semiquantitative analysis. CONCLUSION The amyloid discordant cases could be overcome by combining CSF Aβ42, CSF ratio, and Amy-PET results. The concordance of any 2 out of the 3 biomarkers seems to reveal the remaining one as a false result. A cut-off point review could avoid CSF Aβ42 false-negative results. The regional semiquantitative Amy-PET analysis in AD areas, such as precuneus and PCC, could increase the accuracy in AD diagnosis.
Collapse
Affiliation(s)
- Matilde Nerattini
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy.
| | - Federica Rubino
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy
| | - Annachiara Arnone
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy
| | - Cristina Polito
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy
| | - Salvatore Mazzeo
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy
| | - Gemma Lombardi
- IRCCS Fondazione Don Carlo Gnocchi, Via Scandicci 269, 50143, Florence, Italy
| | - Giulia Puccini
- Department of Nuclear Medicine, Hospital of Prato, Via Suor Niccolina Infermiera, 20/22, 59100, Prato, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Via Scandicci 269, 50143, Florence, Italy
| | - Maria Teresa De Cristofaro
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Via Scandicci 269, 50143, Florence, Italy
| | - Alberto Pupi
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy
| | - Roberto Sciagrà
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy
| | - Valentina Bessi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy
| | - Valentina Berti
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy
| |
Collapse
|
98
|
Bjorkli C, Louet C, Flo TH, Hemler M, Sandvig A, Sandvig I. In Vivo Microdialysis in Mice Captures Changes in Alzheimer's Disease Cerebrospinal Fluid Biomarkers Consistent with Developing Pathology. J Alzheimers Dis 2021; 84:1781-1794. [PMID: 34719495 DOI: 10.3233/jad-210715] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Preclinical models of Alzheimer's disease (AD) can provide valuable insights into the onset and progression of the disease, such as changes in concentrations of amyloid-β (Aβ) and tau in cerebrospinal fluid (CSF). However, such models are currently underutilized due to limited advancement in techniques that allow for longitudinal CSF monitoring. OBJECTIVE An elegant way to understand the biochemical environment in the diseased brain is intracerebral microdialysis, a method that has until now been limited to short-term observations, or snapshots, of the brain microenvironment. Here we draw upon patient-based findings to characterize CSF biomarkers in a commonly used preclinical mouse model for AD. METHODS Our modified push-pull microdialysis method was first validated ex vivo with human CSF samples, and then in vivo in an AD mouse model, permitting assessment of dynamic changes of CSF Aβ and tau and allowing for better translational understanding of CSF biomarkers. RESULTS We demonstrate that CSF biomarker changes in preclinical models capture what is observed in the brain; with a decrease in CSF Aβ observed when plaques are deposited, and an increase in CSF tau once tau pathology is present in the brain parenchyma. We found that a high molecular weight cut-off membrane allowed for simultaneous sampling of Aβ and tau, comparable to CSF collection by lumbar puncture in patients. CONCLUSION Our approach can further advance AD and other neurodegenerative research by following evolving neuropathology along the disease cascade via consecutive sampling from the same animal and can additionally be used to administer pharmaceutical compounds and assess their efficacy (Bjorkli, unpublished data).
Collapse
Affiliation(s)
- Christiana Bjorkli
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Claire Louet
- Center for Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Trude Helen Flo
- Center for Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Mary Hemler
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Axel Sandvig
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Clinical Neuroscience, Neuro, Head and Neck, Umeå University Hospital, Umeå, Sweden.,Department of Community Medicine and Rehabilitation, Neuro, Head and Neck, Umeå University Hospital, Umeå, Sweden
| | - Ioanna Sandvig
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
99
|
Karami A, Darreh-Shori T, Schultzberg M, Eriksdotter M. CSF and Plasma Cholinergic Markers in Patients With Cognitive Impairment. Front Aging Neurosci 2021; 13:704583. [PMID: 34512307 PMCID: PMC8426513 DOI: 10.3389/fnagi.2021.704583] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/21/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction Alzheimer’s disease (AD) is the most prevalent form of dementia with symptoms of deteriorating cognitive functions and memory loss, partially as a result of a decrease in cholinergic neurotransmission. The disease is incurable and treatment with cholinesterase inhibitors (ChEIs) is symptomatic. Choline acetyltransferase (ChAT), the enzyme that synthesizes acetylcholine (ACh), has been proven recently to be present in both cerebrospinal fluid (CSF) and plasma. As ChAT plays a role in regulating the extracellular ACh levels, it may have an impact on prognosis and cognitive performance in AD patients. Objectives To measure ChAT activity and its protein concentration in CSF and plasma from patients with AD, mild cognitive impairment (MCI), or Subjective cognitive impairment (SCI). Methods Plasma and CSF samples were obtained from 21 AD, 32 MCI, and 30 SCI patients. The activity and protein levels of ChAT and acetylcholinesterase (AChE), the enzyme catalyzing the hydrolysis of ACh, were analyzed using an integrated activity and protein concentration ELISA-like assay. A Cholinergic Index was calculated as the ratio of ChAT to AChE activities in CSF. The data were analyzed in relation to dementia biomarkers and cognitive performance of the patients. Results The CSF ChAT activity was significantly higher (55–67%) in MCI patients compared to AD and SCI cases. The CSF Cholinergic Index was 41 and 22% lower in AD patients than in MCI and SCI subjects, respectively. This index correlated positively with the Aβ42/p-tau ratio in CSF in SCI but negatively with that in AD and MCI. The ChAT activity and protein levels in plasma exhibited significant differences with the pattern of AD>>MCI>SCI. Conclusion This is the first study investigating soluble levels of the key cholinergic enzyme, ChAT, in both plasma and CSF of individuals at different clinical stages of dementia. Although further validation is needed, the overall pattern of the results suggests that in the continuum of AD, the cholinergic signaling exhibits an inverse U-shape dynamic of changes in the brain that greatly differs from the changes observed in the plasma compartment.
Collapse
Affiliation(s)
- Azadeh Karami
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Taher Darreh-Shori
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Marianne Schultzberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Campus Solna, Stockholm, Sweden
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden.,Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
100
|
Rodrigues PM, Bispo BC, Garrett C, Alves D, Teixeira JP, Freitas D. Lacsogram: A New EEG Tool to Diagnose Alzheimer's Disease. IEEE J Biomed Health Inform 2021; 25:3384-3395. [PMID: 33784628 DOI: 10.1109/jbhi.2021.3069789] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
This work proposes the application of a new electroencephalogram (EEG) signal processing tool - the lacsogram - to characterize the Alzheimer's disease (AD) activity and to assist on its diagnosis at different stages: Mild Cognitive Impairment (MCI), Mild and Moderate AD (ADM) and Advanced AD (ADA). Statistical analyzes are performed to lacstral distances between conventional EEG subbands to find measures capable of discriminating AD in all stages and characterizing the AD activity in each electrode. Cepstral distances are used for comparison. Comparing all AD stages and Controls (C), the most important significances are the lacstral distances between subbands θ and α ( p = 0.0014 0.05). The topographic maps show significant differences in parietal, temporal and frontal regions as AD progresses. Machine learning models with a leave-one-out cross-validation process are applied to lacstral/cepstral distances to develop an automatic method for diagnosing AD. The following classification accuracies are obtained with an artificial neural network: 95.55% for All vs All, 98.06% for C vs MCI, 95.99% for C vs ADM, 93.85% for MCI vs ADM-ADA. In C vs MCI, C vs ADM and MCI vs ADM-ADA, the proposed method outperforms the state-of-art methods by 5%, 1%, and 2%, respectively. In All vs All, it outperforms the state-of-art EEG and non-EEG methods by 6% and 2%, respectively. These results indicate that the proposed method represents an improvement in diagnosing AD.
Collapse
|