1
|
Bouteloup V, Villain N, Vidal JS, Gonzalez-Ortiz F, Yuksekel I, Santos C, Schraen-Maschken S, Pellegrin I, Lehmann S, Blennow K, Chêne G, Hanon O, Dufouil C, Planche V. Cognitive Phenotyping and Interpretation of Alzheimer Blood Biomarkers. JAMA Neurol 2025:2831276. [PMID: 40181683 PMCID: PMC11971688 DOI: 10.1001/jamaneurol.2025.0142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/16/2025] [Indexed: 04/05/2025]
Abstract
Importance Blood phosphorylated tau 217 (p-tau217) showed good performance in predicting brain amyloidosis. However, the importance of detailed cognitive phenotyping in patients without dementia when interpreting p-tau217 results remains unclear. Objective To assess whether accuracy, negative predictive value (NPV), and positive predictive value (PPV) in predicting brain amyloidosis using p-tau217 varies across clinical presentations in patients without dementia. Design, Setting, and Participants The study design included 2 observational, prospective cohort studies: The Cohort of Outpatients From French Research Memory Centers in Order to Improve Knowledge on Alzheimer's Disease and Related Disorders (MEMENTO), with enrollment from 2011 to 2014 and 5 years of follow-up, and the Biomarker of Amyloid Peptide and Alzheimer's Disease Risk (BALTAZAR) cohort study, with enrollment from 2010 to 2015 and 3 years of follow-up. Both are multicenter cohorts conducted in French memory clinics. Participants without dementia were included for analysis if they had baseline blood p-tau217 measurement and a known amyloid status through cerebrospinal fluid amyloid β (Aβ)-42/Aβ-40 ratio or positron emission tomography. They presented with either subjective cognitive impairment (SCI), mild cognitive impairment (MCI) with a common Alzheimer disease (AD) phenotype (cAD-MCI: amnestic syndrome of hippocampal type, posterior cortical atrophy, or logopenic primary progressive aphasia), or MCI with uncommon AD or other phenotypes (uAD-MCI). Data were analyzed from May to September 2024. Exposures Blood p-tau217 concentrations. Main Outcomes and Measures Brain amyloidosis probabilities were derived from p-tau217 logistic regressions including age, gender, and APOE genotype. Published and internally developed cut points with 90% sensitivity and specificity were used. Results A total of 776 participants from the MEMENTO cohort (N = 2323 participants) and 193 participants from the BALTAZAR cohort (N = 1040) were included in this analysis. In the MEMENTO cohort (median [IQR] age, 71 [65-76] years; 444 female [57%]), brain amyloidosis prevalence was 16.5% (20 of 121) in SCI, 45.9% (78 of 170) in cAD-MCI, and 24.5% (119 of 485) in uAD-MCI. Area under the receiver operating characteristic curve for predicting brain amyloidosis with p-tau217 models was 0.78 (95% CI, 0.66-0.89), 0.91 (95% CI, 0.86-0.95), and 0.87 (95% CI, 0.84-0.91) in the SCI, cAD-MCI, and uAD-MCI subgroups, respectively. External cut points resulted in a PPV of 60.0%, 90.0%, and 74.5% in the SCI, cAD-MCI, and uAD-MCI subgroups, respectively. NPV ranged from 84.2% to 90.2%. With internally developed cut points, PPVs were 52.6%, 84.0%, and 72.3% in the SCI, cAD-MCI, and uAD-MCI subgroups, respectively. NPVs were high (91.7%-94.6%) in all subgroups. Rates of incident dementia strongly increased with the probability of brain amyloidosis in the cAD-MCI subgroup. Replicated analyses in the BALTAZAR cohort provided similar results. Conclusions and Relevance Results from 2 clinical cohorts suggest that amyloid prevalence varied across cognitive phenotypes and was associated with the diagnostic performance of blood p-tau217 models to determine brain amyloidosis. Comprehensive cognitive phenotyping beyond the basic characterization of SCI, MCI, or dementia should accompany the use of blood biomarkers in clinical practice to avoid misdiagnosis due to false positives.
Collapse
Affiliation(s)
- Vincent Bouteloup
- Bordeaux Population Health, University of Bordeaux, Inserm, UMR1219, Bordeaux, France
- CIC 1401 EC, Pôle Santé Publique, CHU de Bordeaux, Bordeaux, France
| | - Nicolas Villain
- Sorbonne Université, INSERM U1127, CNRS 7225, Institut du Cerveau–ICM, Paris, France
- Department of Neurology, Institute of Memory and Alzheimer’s Disease, AP-HP Sorbonne Université, Pitié-Salpêtrière Hospital, Paris, France
| | - Jean Sebastien Vidal
- Memory Resource and Research Centre of Paris-Broca-Ile de France, APHP, Hôpitaux Universitaires Paris Centre, Hospital Broca, Paris, France
- Université Paris Cité, EA 4468, Paris, France
| | - Fernando Gonzalez-Ortiz
- Inst. of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Lab, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Idil Yuksekel
- Sorbonne Université, INSERM U1127, CNRS 7225, Institut du Cerveau–ICM, Paris, France
| | - Cristiano Santos
- Clinical Neurochemistry Lab, Sahlgrenska University Hospital, Mölndal, Sweden
| | | | - Isabelle Pellegrin
- Laboratory of Immunology and Immunogenetics, Resources Biological Center, CHU Bordeaux, Bordeaux, France
- Univ. Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, Bordeaux, France
| | - Sylvain Lehmann
- LBPC-PPC, Univ Montpellier, INM INSERM, CHU Montpellier, Montpellier, France
| | - Kaj Blennow
- Sorbonne Université, INSERM U1127, CNRS 7225, Institut du Cerveau–ICM, Paris, France
- Inst. of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Lab, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Geneviève Chêne
- Bordeaux Population Health, University of Bordeaux, Inserm, UMR1219, Bordeaux, France
- CIC 1401 EC, Pôle Santé Publique, CHU de Bordeaux, Bordeaux, France
| | - Olivier Hanon
- Memory Resource and Research Centre of Paris-Broca-Ile de France, APHP, Hôpitaux Universitaires Paris Centre, Hospital Broca, Paris, France
- Université Paris Cité, EA 4468, Paris, France
| | - Carole Dufouil
- Bordeaux Population Health, University of Bordeaux, Inserm, UMR1219, Bordeaux, France
- CIC 1401 EC, Pôle Santé Publique, CHU de Bordeaux, Bordeaux, France
| | - Vincent Planche
- Institut des Maladies Neurodégénératives, Univ. Bordeaux, CNRS, UMR 5293, Bordeaux, France
- Pôle de Neurosciences Cliniques, Centre Mémoire de Ressources et de Recherche, CHU de Bordeaux, Bordeaux, France
| |
Collapse
|
2
|
Lalive HM, Griffa A, Pini L, Rouaud O, Allali G. Biomarkers do not paint the whole picture: The role of clinical expertise and advanced neuroimaging for Alzheimer's disease diagnosis. J Alzheimers Dis 2025:13872877251328953. [PMID: 40111924 DOI: 10.1177/13872877251328953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Accurate diagnosis of Alzheimer's disease (AD) in Memory Clinics remains challenging due to the limited specificity of conventional clinical assessment and structural imaging. The recent commentary by Vyhnalek and colleagues advocates for the incorporation of molecular biomarkers for AD diagnosis in clinical practice. However, this approach only partially captures the complexity of disease expression due to co-pathologies such as limbic-predominant age-related TDP-43 encephalopathy, a mimic of AD. At the era of immunotherapy for AD, clinical expertise remains essential to identify AD from its mimics, especially when both entities co-exist, and may rely on advanced neuroimaging techniques such as brain connectivity.
Collapse
Affiliation(s)
- Hadrien M Lalive
- Department of Clinical Neurosciences, Leenaards Memory Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Alessandra Griffa
- Department of Clinical Neurosciences, Leenaards Memory Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Medical Image Processing Laboratory, Neuro-X Institute, École Polytechnique Fédérale De Lausanne (EPFL), Geneva, Switzerland
| | - Lorenzo Pini
- Department of Neuroscience, University of Padova, Padova, Italy
| | - Olivier Rouaud
- Department of Clinical Neurosciences, Leenaards Memory Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Gilles Allali
- Department of Clinical Neurosciences, Leenaards Memory Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
3
|
Coughlan GT, Klinger HM, Boyle R, Betthauser TJ, Binette AP, Christenson L, Chadwick T, Hansson O, Harrison TM, Healy B, Jacobs HIL, Hanseeuw B, Jonaitis E, Jack CR, Johnson KA, Langhough RE, Properzi MJ, Rentz DM, Schultz AP, Smith R, Seto M, Johnson SC, Mielke MM, Shirzadi Z, Yau WYW, Manson JE, Sperling RA, Vemuri P, Buckley RF. Sex Differences in Longitudinal Tau-PET in Preclinical Alzheimer Disease: A Meta-Analysis. JAMA Neurol 2025:2830857. [PMID: 40029638 DOI: 10.1001/jamaneurol.2025.0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Importance Alzheimer disease (AD) predominates in females at almost twice the rate relative to males. Mounting evidence in adults without AD indicates that females exhibit higher tau deposition than age-matched males, particularly in the setting of elevated β-amyloid (Aβ), but the evidence for sex differences in tau accumulation rates is inconclusive. Objective To examine whether female sex is associated with faster tau accumulation in the setting of high Aβ (as measured with positron emission tomography [PET]) and the moderating influence of sex on the association between APOEε4 carrier status and tau accumulation. Data Sources This meta-analysis used data from 6 longitudinal aging and AD studies, including the Alzheimer's Disease Neuroimaging Initiative, Berkeley Aging Cohort Study, BioFINDER 1, Harvard Aging Brain Study, Mayo Clinic Study of Aging, and Wisconsin Registry for Alzheimer Prevention. Longitudinal data were collected between November 2004 and May 2022. Study Selection Included studies required available longitudinal [18F]flortaucipir or [18F]-MK-6240 tau-PET scans, as well as baseline [11C] Pittsburgh Compound B, [18F]flutemetamol or [18F]florbetapir Aβ-PET scans. Recruitment criteria varied across studies. Analyses began on August 7, 2023, and were completed on February 5, 2024. Data Extraction and Synthesis In each study, primary analyses extracted estimates for the sex (female or male) and the sex by baseline Aβ-PET status (high or low) association with longitudinal tau-PET using a series of mixed-effects models. Secondary mixed-effects models extracted the interaction estimate for the association of sex by APOEε4 carrier status with longitudinal tau-PET. Study-specific estimates for each mixed-effects model were then pooled in a meta-analysis, and the global fixed effect (β) and total heterogeneity (I2) across studies were estimated. This study is reported following the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) reporting guideline. Main Outcomes and Measures Seven tau-PET outcomes that showed cross-sectional sex differences were examined across temporal, parietal, and occipital lobes. Results Among 6 studies assessed, there were 1376 participants (761 [55%] female; mean [range] age at first tau scan, 71.9 [46-93] years; 401 participants [29%] with high baseline Aβ; 412 APOEε4 carriers [30%]). Among individuals with high baseline Aβ, female sex was associated with faster tau accumulation localized to inferior temporal (β = -0.14; 95% CI, -0.22 to -0.06; P = .009) temporal fusiform (β = -0.13; 95% CI, -0.23 to -0.04; P = .02), and lateral occipital regions (β = -0.15; 95% CI, -0.24 to -0.06; P = .009) compared with male sex. Among APOEε4 carriers, female sex was associated with faster inferior-temporal tau accumulation (β = -0.10; 95% CI, -0.16 to -0.03; P = .01). Conclusions and Relevance These findings suggest that sex differences in the pathological progression of AD call for sex-specific timing considerations when administrating anti-Aβ and anti-tau treatments.
Collapse
Affiliation(s)
- Gillian T Coughlan
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Hannah M Klinger
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Rory Boyle
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Tobey J Betthauser
- Department of Medicine, Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden and Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Luke Christenson
- Department of Radiology, Mayo Clinic Rochester, Rochester, Minnesota
| | - Trevor Chadwick
- Department of Neuroscience, University of California, Berkeley
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden and Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | | | - Brian Healy
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Heidi I L Jacobs
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Bernard Hanseeuw
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
- Department of Neurology, Cliniques Universitaires Saint-Luc, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Erin Jonaitis
- Department of Medicine, Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic Rochester, Rochester, Minnesota
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Rebecca E Langhough
- Department of Medicine, Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison
| | - Michael J Properzi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Dorene M Rentz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden and Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Mabel Seto
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Sterling C Johnson
- Department of Medicine, Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison
| | - Michelle M Mielke
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Zahra Shirzadi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Wai-Ying Wendy Yau
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - JoAnn E Manson
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, and the Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Prashanthi Vemuri
- Department of Radiology, Mayo Clinic Rochester, Rochester, Minnesota
| | - Rachel F Buckley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
4
|
Soncu Büyükişcan E. Neuropsychology of Alzheimer's disease: From preclinical phase to dementia. APPLIED NEUROPSYCHOLOGY. ADULT 2025:1-9. [PMID: 39982692 DOI: 10.1080/23279095.2025.2469236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by significant cognitive and functional decline, initially presenting with episodic memory impairment. A thorough neuropsychological assessment is essential for AD diagnosis, particularly in the early stages in which interventions may be more effective. This paper reviews the neuropsychology of Alzheimer's disease, highlighting the cognitive progression of the disease. In the typical forms of AD, episodic memory appears to be the first and foremost affected cognitive domain. As AD progresses, cognitive impairments extend beyond memory to affect various domains such as attention, executive functions, language, and visuospatial abilities. Neuropsychiatric issues, such as depression and anxiety, which often accompany cognitive decline, are also common, especially at the advanced stages of the disease. While episodic memory impairment is the earliest and most prominent feature in typical AD cases, comprehensive assessments, including social cognition and neuropsychiatric evaluations, are crucial for accurate diagnosis and treatment planning.
Collapse
|
5
|
Park J, Kim WJ, Jung HW, Kim JJ, Park JY. Relationship between regional relative theta power and amyloid deposition in mild cognitive impairment: an exploratory study. Front Neurosci 2025; 19:1510878. [PMID: 39991752 PMCID: PMC11842361 DOI: 10.3389/fnins.2025.1510878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/22/2025] [Indexed: 02/25/2025] Open
Abstract
Introduction Electroencephalographic (EEG) abnormalities, such as increased theta power, have been proposed as biomarkers for neurocognitive disorders. Advancements in amyloid positron emission tomography (PET) imaging have enhanced our understanding of the pathology of neurocognitive disorders, such as amyloid deposition. However, much remains unknown regarding the relationship between regional amyloid deposition and EEG abnormalities. This study aimed to explore the relationship between regional EEG abnormalities and amyloid deposition in patients with mild cognitive impairment (MCI). Methods We recruited 24 older adults with MCI from a community center for dementia prevention, and 21 participants were included in the final analysis. EEG was recorded using a 64-channel system, and amyloid deposition was measured using amyloid PET imaging. Magnetic resonance imaging (MRI) data were used to create individualized brain models for EEG source localization. Correlations between relative theta power and standardized uptake value ratios (SUVRs) in 12 brain regions were analyzed using Spearman's correlation coefficient. Results Significant positive correlations between relative theta power and SUVR values were found in several brain regions in the individualized brain model during the resting eyes-closed condition, including right temporal lobe (r = 0.581, p = 0.006), left hippocampus (r = 0.438, p = 0.047), left parietal lobe (r = 0.471, p = 0.031), right parietal lobe (r = 0.509, p = 0.018), left occipital lobe (r = 0.597, p = 0.004), and right occipital lobe (r = 0.590, p = 0.005). During the visual working memory condition, significant correlations were found in both cingulate lobes (left: r = 0.483, p = 0.027; right: r = 0.449, p = 0.041), left parietal lobe (r = 0.530, p = 0.010), right parietal lobe (r = 0.606, p = 0.004), left occipital lobe (r = 0.648, p = 0.001), and right occipital lobe (r = 0.657, p = 0.001). Conclusion The result suggests that regional increases in relative theta power are associated with regional amyloid deposition in patients with MCI. These findings highlight the potential of EEG in detecting amyloid deposition. Future large-scale studies are needed to validate these preliminary findings and explore their clinical applications.
Collapse
Affiliation(s)
- Jaesub Park
- Department of Psychiatry, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Gyeonggi, Republic of Korea
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Woo Jung Kim
- Department of Psychiatry, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Gyeonggi, Republic of Korea
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Han Wool Jung
- Department of Psychiatry, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Gyeonggi, Republic of Korea
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae-Jin Kim
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Psychiatry, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin Young Park
- Department of Psychiatry, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Gyeonggi, Republic of Korea
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Center for Digital Health, Yongin Severance Hospital, Yonsei University Health System, Yongin, Gyeonggi, Republic of Korea
| |
Collapse
|
6
|
Rabinovici GD, Knopman DS, Arbizu J, Benzinger TLS, Donohoe KJ, Hansson O, Herscovitch P, Kuo PH, Lingler JH, Minoshima S, Murray ME, Price JC, Salloway SP, Weber CJ, Carrillo MC, Johnson KA. Updated Appropriate Use Criteria for Amyloid and Tau PET: A Report from the Alzheimer's Association and Society for Nuclear Medicine and Molecular Imaging Workgroup. J Nucl Med 2025:jnumed.124.268756. [PMID: 39778970 DOI: 10.2967/jnumed.124.268756] [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: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 01/11/2025] Open
Abstract
The Alzheimer's Association and the Society of Nuclear Medicine and Molecular Imaging convened a multidisciplinary workgroup to update appropriate use criteria (AUC) for amyloid positron emission tomography (PET) and to develop AUC for tau PET. Methods: The workgroup identified key research questions that guided a systematic literature review on clinical amyloid/tau PET. Building on this review, the workgroup developed 17 clinical scenarios in which amyloid or tau PET may be considered. A modified Delphi approach was used to rate each scenario by consensus as "rarely appropriate," "uncertain," or "appropriate." Ratings were performed separately for amyloid and tau PET as stand-alone modalities. Results: For amyloid PET, 7 scenarios were rated as appropriate, 2 as uncertain, and 8 as rarely appropriate. For tau PET, 5 scenarios were rated as appropriate, 6 as uncertain, and 6 as rarely appropriate. Conclusion: AUC for amyloid and tau PET provide expert recommendations for clinical use of these technologies in the evolving landscape of diagnostics and therapeutics for Alzheimer's disease.
Collapse
Affiliation(s)
- Gil D Rabinovici
- Department of Neurology and Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California;
| | - David S Knopman
- Mayo Clinic Neurology and Neurosurgery, Rochester, Minnesota
| | - Javier Arbizu
- Department of Nuclear Medicine, University of Navarra Clinic, Pamplona, Spain
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri; Knight Alzheimer's Disease Research Center, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Kevin J Donohoe
- Nuclear Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Peter Herscovitch
- Positron Emission Tomography Department, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Phillip H Kuo
- Medical Imaging, Medicine, and Biomedical Engineering, University of Arizona, Tucson, Arizona
| | - Jennifer H Lingler
- Department of Health and Community Systems, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Satoshi Minoshima
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah
| | | | - Julie C Price
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Stephen P Salloway
- Department of Neurology and Psychiatry the Warren Alpert School of Medicine, Brown University, Providence, Rhode Island
- Butler Hospital Memory and Aging Program, Providence, Rhode Island
| | | | | | - Keith A Johnson
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
- Molecular Neuroimaging, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts; and
- Departments of Neurology and Radiology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
7
|
Rabinovici GD, Knopman DS, Arbizu J, Benzinger TLS, Donohoe KJ, Hansson O, Herscovitch P, Kuo PH, Lingler JH, Minoshima S, Murray ME, Price JC, Salloway SP, Weber CJ, Carrillo MC, Johnson KA. Updated appropriate use criteria for amyloid and tau PET: A report from the Alzheimer's Association and Society for Nuclear Medicine and Molecular Imaging Workgroup. Alzheimers Dement 2025; 21:e14338. [PMID: 39776249 PMCID: PMC11772739 DOI: 10.1002/alz.14338] [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: 07/19/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION The Alzheimer's Association and the Society of Nuclear Medicine and Molecular Imaging convened a multidisciplinary workgroup to update appropriate use criteria (AUC) for amyloid positron emission tomography (PET) and to develop AUC for tau PET. METHODS The workgroup identified key research questions that guided a systematic literature review on clinical amyloid/tau PET. Building on this review, the workgroup developed 17 clinical scenarios in which amyloid or tau PET may be considered. A modified Delphi approach was used to rate each scenario by consensus as "rarely appropriate," "uncertain," or "appropriate." Ratings were performed separately for amyloid and tau PET as stand-alone modalities. RESULTS For amyloid PET, seven scenarios were rated as appropriate, two as uncertain, and eight as rarely appropriate. For tau PET, five scenarios were rated as appropriate, six as uncertain, and six as rarely appropriate. DISCUSSION AUC for amyloid and tau PET provide expert recommendations for clinical use of these technologies in the evolving landscape of diagnostics and therapeutics for Alzheimer's disease. HIGHLIGHTS A multidisciplinary workgroup convened by the Alzheimer's Association and the Society of Nuclear Medicine and Molecular Imaging updated the appropriate use criteria (AUC) for amyloid positron emission tomography (PET) and to develop AUC for tau PET. The goal of these updated AUC is to assist clinicians in identifying clinical scenarios in which amyloid or tau PET may be useful for guiding the diagnosis and management of patients who have, or are at risk for, cognitive decline These updated AUC are intended for dementia specialists who spend a significant proportion of their clinical effort caring for patients with cognitive complaints, as well as serve as a general reference for a broader audience interested in implementation of amyloid and tau PET in clinical practice.
Collapse
Affiliation(s)
- Gil D. Rabinovici
- Department of Neurology and Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | | | - Javier Arbizu
- Department of Nuclear MedicineUniversity of Navarra ClinicPamplonaSpain
| | - Tammie L. S. Benzinger
- Mallinckrodt Institute of RadiologyWashington University in St. Louis School of MedicineSt. LouisMissouriUSA
- Knight Alzheimer's Disease Research CenterWashington University in St. Louis School of MedicineSt. LouisMissouriUSA
| | - Kevin J. Donohoe
- Nuclear Medicine, Beth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Oskar Hansson
- Department of Clinical Sciences MalmöClinical Memory Research UnitFaculty of MedicineLund UniversityLundSweden
- Memory Clinic, Skåne University HospitalSkånes universitetssjukhusMalmöSweden
| | - Peter Herscovitch
- Positron Emission Tomography DepartmentNational Institutes of Health Clinical CenterBethesdaMarylandUSA
| | - Phillip H. Kuo
- Medical Imaging, Medicine, and Biomedical EngineeringUniversity of ArizonaTucsonArizonaUSA
| | - Jennifer H. Lingler
- Department of Health and Community SystemsUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Satoshi Minoshima
- Department of Radiology and Imaging SciencesUniversity of UtahSalt Lake CityUtahUSA
| | | | - Julie C. Price
- Department of RadiologyMassachusetts General Hospital, BostonCharlestownMassachusettsUSA
| | - Stephen P. Salloway
- Department of Neurology and Psychiatry the Warren Alpert School of Medicine at Brown UniversityProvidenceRhode IslandUSA
- Butler Hospital Memory and Aging ProgramProvidenceRhode IslandUSA
| | | | - Maria C. Carrillo
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
| | - Keith A. Johnson
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Molecular Neuroimaging, Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
- Departments of Neurology and RadiologyMassachusetts General HospitalBostonMassachusettsUSA
| |
Collapse
|
8
|
Wang C, Zhou L, Zhou F, Fu T. The application value of Rs-fMRI-based machine learning models for differentiating mild cognitive impairment from Alzheimer's disease: a systematic review and meta-analysis. Neurol Sci 2025; 46:45-62. [PMID: 39225837 PMCID: PMC11698789 DOI: 10.1007/s10072-024-07731-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Various machine learning (ML) models based on resting-state functional MRI (Rs-fMRI) have been developed to facilitate differential diagnosis of mild cognitive impairment (MCI) and Alzheimer's disease (AD). However, the diagnostic accuracy of such models remains understudied. Therefore, we conducted this systematic review and meta-analysis to explore the diagnostic accuracy of Rs-fMRI-based radiomics in differentiating MCI from AD. METHODS PubMed, Embase, Cochrane, and Web of Science were searched from inception up to February 8, 2024, to identify relevant studies. Meta-analysis was conducted using a bivariate mixed-effects model, and sub-group analyses were carried out by the types of ML tasks (binary classification and multi-class classification tasks). FINDINGS In total, 23 studies, comprising 5,554 participants were enrolled in the study. In the binary classification tasks (twenty studies), the diagnostic accuracy of the ML model for AD was 0.99 (95%CI: 0.34 ~ 1.00), with a sensitivity of 0.94 (95%CI: 0.89 ~ 0.97) and a specificity of 0.98 (95%CI: 0.95 ~ 1.00). In the multi-class classification tasks (six studies), the diagnostic accuracy of the ML model was 0.98 (95%CI: 0.98 ~ 0.99) for NC, 0.96 (95%CI: 0.96 ~ 0.96) for early mild cognitive impairment (EMCI), 0.97 (95%CI: 0.96 ~ 0.97) for late mild cognitive impairment (LMCI), and 0.95 (95%CI: 0.95 ~ 0.95) for AD. CONCLUSIONS The Rs-fMRI-based ML model can be adapted to multi-class classification tasks. Therefore, multi-center studies with large samples are needed to develop intelligent application tools to promote the development of intelligent ML models for disease diagnosis.
Collapse
Affiliation(s)
- Chentong Wang
- Rheumatology Immunology Department, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, 315000, China
| | - Li Zhou
- Rheumatology Immunology Department, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, 315000, China.
- Ningbo Medical Center Lihuili Hospital, 1111 Jiangnan Road, Yinzhou District, Ningbo, Zhejiang, China.
| | - Feng Zhou
- Rheumatology Immunology Department, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, 315000, China
| | - Tingting Fu
- Rheumatology Immunology Department, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, 315000, China
| |
Collapse
|
9
|
Raposo Pereira F, Chaumon M, Dubois B, Bakardjian H, Bahrami M, Habert MO, Andrade K, Younsi N, La Corte V, George N. Recognition memory decline is associated with the progression to prodromal Alzheimer's disease in asymptomatic at-risk individuals. J Neurol 2024; 272:70. [PMID: 39680203 DOI: 10.1007/s00415-024-12834-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 12/17/2024]
Abstract
Episodic memory (EM) alterations are a hallmark of Alzheimer's disease (AD). We assessed EM longitudinally in cognitively normal elders at-risk for AD (with subjective memory complaints), as a function of amyloid-β (Aβ) burden, neurodegeneration (N), and progression to prodromal AD. We stratified 264 INSIGHT-preAD study subjects in controls (Aβ-/N-), stable/N- or N + (Aβ +), and progressors/N- or N + (Aβ +) groups (progressors were included only until AD-diagnosis). We used linear mixed-effect models with Aβ and N status, or progression to AD as factors, to analyze behavioral performance in an old/new word-recognition task based on the free and cued selective reminding test (FCSRT). The controls and stable/N- groups showed near-ceiling accuracy and RT improvement across follow-up. The stable/N + group showed accuracy reduction and no RT improvement, i.e., Aβ + /N + cumulative effect. The progressors showed a marked performance decline. EM alterations may constitute early preclinical markers of progression to prodromal AD, while individuals are cognitively normal according to neuropsychological standards.
Collapse
Affiliation(s)
- Filipa Raposo Pereira
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, INSERM, U 1127, CNRS, UMR 7225, AP-HP, CENIR, Centre MEG-EEG, Hôpital de La Pitié-Salpêtrière, 47 Boulevard de L'Hôpital, 75013, Paris, France.
- Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière University Hospital, AP-HP, Paris, France.
| | - Maximilien Chaumon
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, INSERM, U 1127, CNRS, UMR 7225, AP-HP, CENIR, Centre MEG-EEG, Hôpital de La Pitié-Salpêtrière, 47 Boulevard de L'Hôpital, 75013, Paris, France
| | - Bruno Dubois
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, INSERM, U 1127, CNRS, UMR 7225, AP-HP, CENIR, Centre MEG-EEG, Hôpital de La Pitié-Salpêtrière, 47 Boulevard de L'Hôpital, 75013, Paris, France
- Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière University Hospital, AP-HP, Paris, France
- Department of Neurology, Centre of Excellence of Neurodegenerative Disease (CoEN), ICM, CIC Neurosciences, APHP, Hopital Pitie-Salpetriere, Sorbonne Universite, Paris, France
| | - Hovagim Bakardjian
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, INSERM, U 1127, CNRS, UMR 7225, AP-HP, CENIR, Centre MEG-EEG, Hôpital de La Pitié-Salpêtrière, 47 Boulevard de L'Hôpital, 75013, Paris, France
- Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière University Hospital, AP-HP, Paris, France
| | - Mahsa Bahrami
- Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière University Hospital, AP-HP, Paris, France
| | - Marie-Odile Habert
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB, 75006, Paris, France
- AP-HP, Hôpital de la Pitié-Salpêtrière, Médecine Nucléaire, 75013, Paris, France
- Centre d'Acquisition et Traitement des Images (CATI), Paris, France
| | - Katia Andrade
- Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière University Hospital, AP-HP, Paris, France
| | - Nadjia Younsi
- Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière University Hospital, AP-HP, Paris, France
| | - Valentina La Corte
- Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière University Hospital, AP-HP, Paris, France
- Laboratoire Mémoire Cerveau et Cognition (MC2Lab) EA 7536, Institut de Psychologie, Université Paris Cité, Boulogne-Billancourt, France
- Institut Universitaire de France, Paris, France
| | - Nathalie George
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, INSERM, U 1127, CNRS, UMR 7225, AP-HP, CENIR, Centre MEG-EEG, Hôpital de La Pitié-Salpêtrière, 47 Boulevard de L'Hôpital, 75013, Paris, France
| |
Collapse
|
10
|
Costanzo M, Cutrona C, Leodori G, Malimpensa L, D'antonio F, Conte A, Belvisi D. Exploring easily accessible neurophysiological biomarkers for predicting Alzheimer's disease progression: a systematic review. Alzheimers Res Ther 2024; 16:244. [PMID: 39497149 PMCID: PMC11533378 DOI: 10.1186/s13195-024-01607-4] [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: 01/25/2024] [Accepted: 10/19/2024] [Indexed: 11/06/2024]
Abstract
Alzheimer disease (AD) remains a significant global health concern. The progression from preclinical stages to overt dementia has become a crucial point of interest for researchers. This paper reviews the potential of neurophysiological biomarkers in predicting AD progression, based on a systematic literature search following PRISMA guidelines, including 55 studies. EEG-based techniques have been predominantly employed, whereas TMS studies are less common. Among the investigated neurophysiological measures, spectral power measurements and event-related potentials-based measures, including P300 and N200 latencies, have emerged as the most consistent and reliable biomarkers for predicting the likelihood of conversion to AD. In addition, TMS-based indices of cortical excitability and synaptic plasticity have also shown potential in assessing the risk of conversion to AD. However, concerns persist regarding the methodological discrepancies among studies, the accuracy of these neurophysiological measures in comparison to established AD biomarkers, and their immediate clinical applicability. Further research is needed to validate the predictive capabilities of EEG and TMS measures. Advancements in this area could lead to cost-effective, reliable biomarkers, enhancing diagnostic processes and deepening our understanding of AD pathophysiology.
Collapse
Affiliation(s)
- Matteo Costanzo
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università 30, Rome, 00185, RM, Italy
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy
| | - Carolina Cutrona
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università 30, Rome, 00185, RM, Italy
| | - Giorgio Leodori
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università 30, Rome, 00185, RM, Italy
- IRCCS Neuromed, Via Atinense 18, Pozzilli, 86077, IS, Italy
| | | | - Fabrizia D'antonio
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università 30, Rome, 00185, RM, Italy
| | - Antonella Conte
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università 30, Rome, 00185, RM, Italy
- IRCCS Neuromed, Via Atinense 18, Pozzilli, 86077, IS, Italy
| | - Daniele Belvisi
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università 30, Rome, 00185, RM, Italy.
- IRCCS Neuromed, Via Atinense 18, Pozzilli, 86077, IS, Italy.
| |
Collapse
|
11
|
Le Scouarnec L, Bouteloup V, van der Veere PJ, van der Flier WM, Teunissen CE, Verberk IMW, Planche V, Chêne G, Dufouil C. Development and assessment of algorithms for predicting brain amyloid positivity in a population without dementia. Alzheimers Res Ther 2024; 16:219. [PMID: 39394180 PMCID: PMC11468062 DOI: 10.1186/s13195-024-01595-5] [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: 07/26/2024] [Accepted: 10/02/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND The accumulation of amyloid-β (Aβ) peptide in the brain is a hallmark of Alzheimer's disease (AD), occurring years before symptom onset. Current methods for quantifying in vivo amyloid load involve invasive or costly procedures, limiting accessibility. Early detection of amyloid positivity in non-demented individuals is crucial for aiding early AD diagnosis and for initiating anti-amyloid immunotherapies at early stages. This study aimed to develop and validate predictive models to identify brain amyloid positivity in non-demented patients, using routinely collected clinical data. METHODS Predictive models for amyloid positivity were developed using data from 853 non-demented participants in the MEMENTO cohort. Amyloid levels were measured potentially repeatedly during study course through Positron Emision Tomography or CerebroSpinal Fluid analysis. The probability of amyloid positivity was modelled using mixed-effects logistic regression. Predictors included demographic information, cognitive assessments, visual brain MRI evaluations of hippocampal atrophy and lobar microbleeds, AD-related blood biomarkers (Aβ42/40 and P-tau181), and ApoE4 status. Models were subjected to internal cross-validation and external validation using data from the Amsterdam Dementia Cohort. Performance also was evaluated in a subsample that met the main criteria of the Appropriate Use Recommendations (AUR) for lecanemab. RESULTS The most effective model incorporated demographic data, cognitive assessments, ApoE status, and AD-related blood biomarkers, achieving AUCs of 0.82 [95%CI 0.81-0.82] in MEMENTO sample and 0.90 [95%CI 0.86-0.94] in the external validation sample. This model significantly outperformed a reference model based solely on demographic and cognitive data, with an AUC difference in MEMENTO of 0.10 [95%CI 0.10-0.11]. A similar model without ApoE genotype achieved comparable discriminatory performance. MRI markers did not improve model performance. Performances in AUR of lecanemab subsample were comparable. CONCLUSION A predictive model integrating demographic, cognitive, and blood biomarker data offers a promising method to help identify amyloid status in non-demented patients. ApoE genotype and brain MRI data were not necessary for strong discriminatory ability, suggesting that ApoE genotyping may be deferred when assessing the risk-benefit ratio of immunotherapies in amyloid-positive patients who desire treatment. The integration of this model into clinical practice could reduce the need for lumbar puncture or PET examinations to confirm amyloid status.
Collapse
Affiliation(s)
- Lisa Le Scouarnec
- Univ. Bordeaux, Bordeaux Population Health, UMR1219, Inserm, Bordeaux, France.
- CIC 1401 de Bordeaux - Module Epidémiologique Clinique / Bâtiment ISPED, Université de Bordeaux, 146, rue Léo Saignat, Bordeaux cedex, CS61292 33076, France.
| | - Vincent Bouteloup
- Univ. Bordeaux, Bordeaux Population Health, UMR1219, Inserm, Bordeaux, France
- CIC 1401 de Bordeaux - Module Epidémiologique Clinique / Bâtiment ISPED, Université de Bordeaux, 146, rue Léo Saignat, Bordeaux cedex, CS61292 33076, France
| | - Pieter J van der Veere
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Department of Epidemiology and Biostatistics, Amsterdam Neuroscience, VU University Medical Center, De Boelelaan 1117, Amsterdam, 1081 HV, the Netherlands
- Amsterdam Neuroscience, De Boelelaan 1117, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Department of Epidemiology and Biostatistics, Amsterdam Neuroscience, VU University Medical Center, De Boelelaan 1117, Amsterdam, 1081 HV, the Netherlands
- Amsterdam Neuroscience, De Boelelaan 1117, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Laboratory Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Inge M W Verberk
- Neurochemistry Laboratory, Laboratory Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Vincent Planche
- Institut des Maladies Neurodégénératives, Univ. Bordeaux, CNRS, UMR 5293, Bordeaux, France
- Pôle de Neurosciences Cliniques, Centre Mémoire de Ressources et de Recherche, CHU de, Bordeaux, France
| | - Geneviève Chêne
- Univ. Bordeaux, Bordeaux Population Health, UMR1219, Inserm, Bordeaux, France
- CIC 1401 de Bordeaux - Module Epidémiologique Clinique / Bâtiment ISPED, Université de Bordeaux, 146, rue Léo Saignat, Bordeaux cedex, CS61292 33076, France
| | - Carole Dufouil
- Univ. Bordeaux, Bordeaux Population Health, UMR1219, Inserm, Bordeaux, France
- CIC 1401 de Bordeaux - Module Epidémiologique Clinique / Bâtiment ISPED, Université de Bordeaux, 146, rue Léo Saignat, Bordeaux cedex, CS61292 33076, France
| |
Collapse
|
12
|
Dan YR, Chiam KH. Discovery of plasma biomarkers related to blood-brain barrier dysregulation in Alzheimer's disease. FRONTIERS IN BIOINFORMATICS 2024; 4:1463001. [PMID: 39429696 PMCID: PMC11487119 DOI: 10.3389/fbinf.2024.1463001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/19/2024] [Indexed: 10/22/2024] Open
Abstract
Introduction Blood-based biomarkers are quantitative, non-invasive diagnostic tools. This study aimed to identify candidate biomarkers for Alzheimer's disease (AD) using publicly available omics datasets, using the hypothesis that with blood-brain barrier dysfunction in AD, brain-synthesized proteins can leak into plasma for detection. Methods Differential abundance results of plasma and brain proteomic datasets were integrated to obtain a list of potential biomarkers. Biological validity was investigated with intercellular communication and gene regulatory analyses on brain single-cell transcriptomics data. Results Five proteins (APOD, B2M, CFH, CLU, and C3) fit biomarker criteria. 4 corresponding transcripts (APOD, B2M, CLU, and C3) were overexpressed in AD astrocytes, mediated by AD-related dysregulations in transcription factors regulating neuroinflammation. Additionally, CLU specifically induced downstream expression of neuronal death genes. Discussion In conclusion, a 5-protein panel is shown to effectively identify AD patients, with evidence of disease specificity and biological validity. Future research should investigate the mechanism of protein leakage through the blood-brain barrier.
Collapse
Affiliation(s)
- Yuet Ruh Dan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Bioinformatics Institute, A*STAR, Singapore, Singapore
| | - Keng-Hwee Chiam
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Bioinformatics Institute, A*STAR, Singapore, Singapore
| |
Collapse
|
13
|
Shi X, Zhang X, Ao JF, Yang M. Correlation between Non-HDL-C/HDL-C and Aβ1-42 levels in cerebral infarction-related cognitive dysfunction. Clin Neurol Neurosurg 2024; 245:108503. [PMID: 39178633 DOI: 10.1016/j.clineuro.2024.108503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 08/26/2024]
Abstract
OBJECTIVE Cerebral infarction treatments are most effective if used early after stroke symptoms occur. Also, early detection is crucial for delaying and improving cognitive impairment. This study investigated the relationship between the ratio of non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol (Non-HDL-C/HDL-C), which reflects the entire burden of the cholesterol transported in atherogenic lipoproteins, and the level of β-amyloid 1-42 (Aβ-1-42), a major component of cerebrovascular amyloid deposits, in peripheral blood and cognitive dysfunction secondary to cerebral infarction. METHODS A total of 83 patients with cerebral infarction admitted to Bozhou People's Hospital between June 2019 and June 2022 were assessed. The patients were divided into two groups based on their Mini-Mental State Scale (MMSE) scores: cognitive dysfunction group (n = 30) and non-cognitive dysfunction group (n = 53). In addition, a control group comprising 34 patients with transient cerebral insufficiency or cerebrovascular stenosis was selected. The groups were compared in terms of various clinical factors, including gender, age, hypertension, hyperlipidemia, lipid indexes, Non-HDL-C/HDL-C, and Aβ1-42 levels. Logistic regression analysis was used to identify the risk factors associated with cognitive dysfunction. RESULTS The results showed that hypertensive patients with cognitive dysfunction secondary to cerebral infarction had a higher proportion of frontal lobe, temporal lobe, and thalamus involvement and lower scores on the MMSE compared to the non-cognitive impairment group and control group (p < 0.05). Additionally, the levels of homocysteine (HCY), Non-HDL-C/HDL-C, and Aβ1-42 in peripheral blood were significantly higher in hypertensive patients with cognitive dysfunction compared to the other two groups (all p < 0.05) and were identified as risk factors for cognitive dysfunction secondary to cerebral infarction. Peripheral blood levels of Non-HDL-C/HDL-C and Aβ1-42 are risk factors for secondary cognitive dysfunction following a cerebral infarction. CONCLUSION These data have important clinical implications for understanding the mechanisms underlying cognitive dysfunction in individuals with cerebrovascular disorders, potentially leading to new early interventions for preventing or treating such diseases.
Collapse
Affiliation(s)
- Xiufang Shi
- The Clinical Laboratory of Bozhou People's Hospital, BoZhou 236800, China.
| | - Xiaoqing Zhang
- The Clinical Laboratory of Bozhou People's Hospital, BoZhou 236800, China.
| | - Jia-Fu Ao
- The Clinical Laboratory of Bozhou People's Hospital, BoZhou 236800, China.
| | - Miao Yang
- The Clinical Laboratory of Bozhou People's Hospital, BoZhou 236800, China.
| |
Collapse
|
14
|
Wang H, Li S, Zhang J, Peng W, Li T, Zhang J. Efficacy of selective serotonin reuptake inhibitors-related antidepressants in Alzheimer's disease: a meta-analysis. Eur J Med Res 2024; 29:438. [PMID: 39210432 PMCID: PMC11360319 DOI: 10.1186/s40001-024-02006-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE To study the effects of selective serotonin reuptake inhibitors (SSRIs) on cognitive functions, mental improvements, and adverse effects in patients with Alzheimer's disease (AD). METHODS Registered in INPLASY (INPLASY202450004), five drugs (citalopram, s-citalopram, quetiapine, olanzapine, and sertraline) were selected as representatives. A comprehensive search was conducted in PubMed, EMBASE, Web of Science, and the Cochrane Library up to May 15, 2024. Search terms were combined using Boolean operators, specifically 'AND' between different categories (e.g., 'Alzheimer's Disease' AND 'SSRIs') and 'OR' within the same category (e.g., 'citalopram OR s-citalopram OR quetiapine OR olanzapine OR sertraline'), to ensure a thorough retrieval of relevant studies. The selection followed rigorous inclusion and exclusion criteria for meta-analysis. RESULTS Fourteen articles from 1118 were selected for meta-analysis. The indicators, including Neuropsychiatric Inventory (NPI), Mini-Mental State Examination (MMSE), Brief Psychiatric Rating Scale (BPRS), and Cornell Scale for Depression in Dementia (CSDD), were used to assess the effects of the drugs on AD treatment. According to the results of NPI, CSDD, BPRS, MMSE, and security assessments, the five antidepressants have significant advantages in AD treatment compared with placebo, while the MMSE of the patient treated with the antidepressants did not show notable changes compared with patients treated only with placebo. Statistical analyses were conducted using Review Manager 5.3, employing random-effects models to account for study heterogeneity and sensitivity analyses to test the robustness of our findings. CONCLUSION This study suggests that SSRI-related antidepressants have great potential values in AD treatment, and further research on the application of SSRI-related antidepressants in AD treatment is necessary.
Collapse
Affiliation(s)
- Haiyan Wang
- Department of Traditional Chinese Medicine, The Second Hospital of Shandong University, 247 Beiyuan St, Jinan, 250033, China
| | - Siyi Li
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Jiwei Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Wei Peng
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250000, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Jianxin Zhang
- Department of Traditional Chinese Medicine, The Second Hospital of Shandong University, 247 Beiyuan St, Jinan, 250033, China.
| |
Collapse
|
15
|
Raposo Pereira F, George N, Dalla Barba G, Dubois B, La Corte V. The memory binding test can anticipate Alzheimer's disease diagnosis at an early preclinical stage: a longitudinal study in the INSIGHTpreAD cohort. Front Aging Neurosci 2024; 16:1414419. [PMID: 39175810 PMCID: PMC11340525 DOI: 10.3389/fnagi.2024.1414419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/12/2024] [Indexed: 08/24/2024] Open
Abstract
Introduction Anticipating the diagnosis of Alzheimer's disease (AD) at an early asymptomatic at-risk stage, where therapeutics can more effectively delay conscious cognitive decline, is currently among the biggest challenges in the field. Herein, we aimed to compare the capacity of the Memory Binding Test (MBT) with the official diagnostic tool, the Free and Cued Selective Reminding Test (FCSRT), to anticipate AD diagnosis at an early preclinical stage based on the associative memory component of MBT (binding), suggested as more sensitive to the emergence of subtle episodic memory (EM) deficits (AD hallmark). Methods We assessed the tests performance longitudinally (over 5 years) in 263 cognitively-normal elderly individuals at risk of AD (>6 months of subjective memory complaints) using linear mixed-effect models controlled for age, sex, and education. We stratified participants in 2 models: amyloid-β (Aβ)/neurodegeneration (N) model, assessing Aβ burden and neurodegeneration effect [3 groups: controls (Aβ-/N-); stable/N- (Aβ+); stable/N+ (Aβ+)]; and the stable/progressors model, assessing progression to prodromal-AD effect [2 groups: stable (Aβ+); progressors (Aβ+)], based on 15 subjects who progressed to AD during follow-up (excluded once diagnosed). Results Aβ burden was associated with significantly less MBT-intrusions, while Aβ burden and neurodegeneration together, with the most. Progression status had a strong negative effect on both tests performance. When compared with the FCSRT, the MBT seems to anticipate diagnosis based on a worst performance in a higher number of scores (including binding) in at least a year. Discussion Anticipation of diagnosis to an asymptomatic at-risk stage, while participants remain cognitively-normal according to FCSRT cut-offs and unaware of objective EM deficits, has the potential to delay the onset of AD-linked cognitive decline by applying promising therapeutics before decline becomes too advanced.
Collapse
Affiliation(s)
- Filipa Raposo Pereira
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, INSERM, U 1127, CNRS, UMR 7225, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
- Department of Neurology, Institute of Memory and Alzheimer’s Disease (IM2A), Pitié-Salpêtrière University Hospital, AP-HP, Paris, France
| | - Nathalie George
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, INSERM, U 1127, CNRS, UMR 7225, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | | | - Bruno Dubois
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, INSERM, U 1127, CNRS, UMR 7225, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
- Department of Neurology, Institute of Memory and Alzheimer’s Disease (IM2A), Pitié-Salpêtrière University Hospital, AP-HP, Paris, France
- Centre of Excellence of Neurodegenerative Disease (CoEN), ICM, CIC Neurosciences, APHP, Department of Neurology, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Valentina La Corte
- Department of Neurology, Institute of Memory and Alzheimer’s Disease (IM2A), Pitié-Salpêtrière University Hospital, AP-HP, Paris, France
- Laboratoire Mémoire Cerveau et Cognition (UR 7536), Institut de Psychologie, Université Paris Cité, Boulogne Billancourt, France
- Institut Universitaire de France, Paris, France
| |
Collapse
|
16
|
Giacomucci G, Mazzeo S, Ingannato A, Crucitti C, Bagnoli S, Padiglioni S, Romano L, Galdo G, Emiliani F, Frigerio D, Ferrari C, Moschini V, Morinelli C, Notarelli A, Sorbi S, Nacmias B, Bessi V. Future perspective and clinical applicability of the combined use of plasma phosphorylated tau 181 and neurofilament light chain in Subjective Cognitive Decline and Mild Cognitive Impairment. Sci Rep 2024; 14:11307. [PMID: 38760423 PMCID: PMC11101654 DOI: 10.1038/s41598-024-61655-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/08/2024] [Indexed: 05/19/2024] Open
Abstract
We aimed to assess diagnostic accuracy of plasma p-tau181 and NfL separately and in combination in discriminating Subjective Cognitive Decline (SCD) and Mild Cognitive Impairment (MCI) patients carrying Alzheimer's Disease (AD) pathology from non-carriers; to propose a flowchart for the interpretation of the results of plasma p-tau181 and NfL. We included 43 SCD, 41 MCI and 21 AD-demented (AD-d) patients, who underwent plasma p-tau181 and NfL analysis. Twenty-eight SCD, 41 MCI and 21 AD-d patients underwent CSF biomarkers analysis (Aβ1-42, Aβ1-42/1-40, p-tau, t-tau) and were classified as carriers of AD pathology (AP+) it they were A+/T+ , or non-carriers (AP-) when they were A-, A+/T-/N-, or A+/T-/N+ according to the A/T(N) system. Plasma p-tau181 and NfL separately showed a good accuracy (AUC = 0.88), while the combined model (NfL + p-tau181) showed an excellent accuracy (AUC = 0.92) in discriminating AP+ from AP- patients. Plasma p-tau181 and NfL results were moderately concordant (Coehn's k = 0.50, p < 0.001). Based on a logistic regression model, we estimated the risk of AD pathology considering the two biomarkers: 10.91% if both p-tau181 and NfL were negative; 41.10 and 76.49% if only one biomarker was positive (respectively p-tau18 and NfL); 94.88% if both p-tau181 and NfL were positive. Considering the moderate concordance and the risk of presenting an underlying AD pathology according to the positivity of plasma p-tau181 and NfL, we proposed a flow chart to guide the combined use of plasma p-tau181 and NfL and the interpretation of biomarker results to detect AD pathology.
Collapse
Affiliation(s)
- Giulia Giacomucci
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3, 50134, Florence, Italy
| | - Salvatore Mazzeo
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3, 50134, Florence, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Assunta Ingannato
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3, 50134, Florence, Italy
| | - Chiara Crucitti
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3, 50134, Florence, Italy
| | - Silvia Bagnoli
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3, 50134, Florence, Italy
| | - Sonia Padiglioni
- Regional Referral Centre for Relational Criticalities - Tuscany Region, University of Florence, Florence, Italy
- Research and Innovation Centre for Dementia-CRIDEM, AOU Careggi, Florence, Italy
| | | | - Giulia Galdo
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3, 50134, Florence, Italy
| | - Filippo Emiliani
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3, 50134, Florence, Italy
| | - Daniele Frigerio
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3, 50134, Florence, Italy
| | - Camilla Ferrari
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3, 50134, Florence, Italy
| | - Valentina Moschini
- SOD Neurologia I, Dipartimento Neuromuscolo-Scheletrico e degli Organi di Senso, AOU Careggi, Florence, Italy
| | - Carmen Morinelli
- SOD Neurologia I, Dipartimento Neuromuscolo-Scheletrico e degli Organi di Senso, AOU Careggi, Florence, Italy
| | - Antonella Notarelli
- Research and Innovation Centre for Dementia-CRIDEM, AOU Careggi, Florence, Italy
- SOD Neurologia I, Dipartimento Neuromuscolo-Scheletrico e degli Organi di Senso, AOU Careggi, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3, 50134, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3, 50134, Florence, Italy.
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy.
| | - Valentina Bessi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3, 50134, Florence, Italy
- Research and Innovation Centre for Dementia-CRIDEM, AOU Careggi, Florence, Italy
| |
Collapse
|
17
|
Kim HH, Kwon MJ, Jo S, Park JE, Kim JW, Kim JH, Kim SE, Kim KW, Han JW. Exploration of neuroanatomical characteristics to differentiate prodromal Alzheimer's disease from cognitively unimpaired amyloid-positive individuals. Sci Rep 2024; 14:10083. [PMID: 38698190 PMCID: PMC11066072 DOI: 10.1038/s41598-024-60843-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 04/28/2024] [Indexed: 05/05/2024] Open
Abstract
Differentiating clinical stages based solely on positive findings from amyloid PET is challenging. We aimed to investigate the neuroanatomical characteristics at the whole-brain level that differentiate prodromal Alzheimer's disease (AD) from cognitively unimpaired amyloid-positive individuals (CU A+) in relation to amyloid deposition and regional atrophy. We included 45 CU A+ participants and 135 participants with amyloid-positive prodromal AD matched 1:3 by age, sex, and education. All participants underwent 18F-florbetaben positron emission tomography and 3D structural T1-weighted magnetic resonance imaging. We compared the standardized uptake value ratios (SUVRs) and volumes in 80 regions of interest (ROIs) between CU A+ and prodromal AD groups using independent t-tests, and employed the least absolute selection and shrinkage operator (LASSO) logistic regression model to identify ROIs associated with prodromal AD in relation to amyloid deposition, regional atrophy, and their interaction. After applying False Discovery Rate correction at < 0.1, there were no differences in global and regional SUVR between CU A+ and prodromal AD groups. Regional volume differences between the two groups were observed in the amygdala, hippocampus, entorhinal cortex, insula, parahippocampal gyrus, and inferior temporal and parietal cortices. LASSO logistic regression model showed significant associations between prodromal AD and atrophy in the entorhinal cortex, inferior parietal cortex, both amygdalae, and left hippocampus. The mean SUVR in the right superior parietal cortex (beta coefficient = 0.0172) and its interaction with the regional volume (0.0672) were also selected in the LASSO model. The mean SUVR in the right superior parietal cortex was associated with an increased likelihood of prodromal AD (Odds ratio [OR] 1.602, p = 0.014), particularly in participants with lower regional volume (OR 3.389, p < 0.001). Only regional volume differences, not amyloid deposition, were observed between CU A+ and prodromal AD. The reduced volume in the superior parietal cortex may play a significant role in the progression to prodromal AD through its interaction with amyloid deposition in that region.
Collapse
Affiliation(s)
- Hak Hyeon Kim
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, 82 Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
- Department of Psychiatry, College of Medicine, Seoul National University, Seoul, South Korea
| | - Min Jeong Kwon
- Department of Brain and Cognitive Science, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Sungman Jo
- Department of Health Science and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Ji Eun Park
- Department of Brain and Cognitive Science, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Ji Won Kim
- Department of Brain and Cognitive Science, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Jae Hyoung Kim
- Department of Radiology, Seoul National University Bundang Hospital, College of Medicine, Seoul National University, Seongnam-si, Gyeonggi-do, South Korea
| | - Sang Eun Kim
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, College of Medicine, Seoul National University, Seongnam-si, Gyeonggi-do, Korea
- Center for Nanomolecular Imaging and Innovative Drug Development, Advanced Institutes of Convergence Technology, Suwon, Republic of Korea
| | - Ki Woong Kim
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, 82 Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
- Department of Psychiatry, College of Medicine, Seoul National University, Seoul, South Korea
- Department of Brain and Cognitive Science, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Department of Health Science and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Ji Won Han
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, 82 Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea.
- Department of Psychiatry, College of Medicine, Seoul National University, Seoul, South Korea.
| |
Collapse
|
18
|
Lopez S, Hampel H, Chiesa PA, Del Percio C, Noce G, Lizio R, Teipel SJ, Dyrba M, González-Escamilla G, Bakardjian H, Cavedo E, Lista S, Vergallo A, Lemercier P, Spinelli G, Grothe MJ, Potier MC, Stocchi F, Ferri R, Habert MO, Dubois B, Babiloni C. The association between posterior resting-state EEG alpha rhythms and functional MRI connectivity in older adults with subjective memory complaint. Neurobiol Aging 2024; 137:62-77. [PMID: 38431999 DOI: 10.1016/j.neurobiolaging.2024.02.008] [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: 05/25/2020] [Revised: 02/09/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
Resting-state eyes-closed electroencephalographic (rsEEG) alpha rhythms are dominant in posterior cortical areas in healthy adults and are abnormal in subjective memory complaint (SMC) persons with Alzheimer's disease amyloidosis. This exploratory study in 161 SMC participants tested the relationships between those rhythms and seed-based resting-state functional magnetic resonance imaging (rs-fMRI) connectivity between thalamus and visual cortical networks as a function of brain amyloid burden, revealed by positron emission tomography and cognitive reserve, measured by educational attainment. The SMC participants were divided into 4 groups according to 2 factors: Education (Edu+ and Edu-) and Amyloid burden (Amy+ and Amy-). There was a statistical interaction (p < 0.05) between the two factors, and the subgroup analysis using estimated marginal means showed a positive association between the mentioned rs-fMRI connectivity and the posterior rsEEG alpha rhythms in the SMC participants with low brain amyloidosis and high CR (Amy-/Edu+). These results suggest that in SMC persons, early Alzheimer's disease amyloidosis may contrast the beneficial effects of cognitive reserve on neurophysiological oscillatory mechanisms at alpha frequencies and connectivity between the thalamus and visual cortical networks.
Collapse
Affiliation(s)
- Susanna Lopez
- Department of Physiology and Pharmacology "Erspamer", Sapienza University of Rome, Rome, Italy
| | - Harald Hampel
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris F-75013, France
| | - Patrizia Andrea Chiesa
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris F-75013, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'hôpital, Paris F-75013, France; Institut du Cerveau et de la Moelle épinière, ICM, INSERM U1127, CNRS UMR 7225, Sorbonne Université, Paris F- 75013, France
| | - Claudio Del Percio
- Department of Physiology and Pharmacology "Erspamer", Sapienza University of Rome, Rome, Italy
| | | | - Roberta Lizio
- Department of Physiology and Pharmacology "Erspamer", Sapienza University of Rome, Rome, Italy
| | - Stefan J Teipel
- Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany; German Center for Neurodegenerative Diseases (DZNE), Greifswald, Rostock, Germany
| | - Martin Dyrba
- Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| | - Gabriel González-Escamilla
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Hovagim Bakardjian
- Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'hôpital, Paris F-75013, France; Centre pour l'Acquisition et le Traitement des Images, (CATI platform), France; Laboratoire d'Imagerie Biomédicale, CNRS, INSERM, Sorbonne University, LIB, Paris F-75006, France
| | - Enrica Cavedo
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris F-75013, France
| | - Simone Lista
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris F-75013, France
| | - Andrea Vergallo
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris F-75013, France
| | - Pablo Lemercier
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris F-75013, France; Centre pour l'Acquisition et le Traitement des Images, (CATI platform), France; Laboratoire d'Imagerie Biomédicale, CNRS, INSERM, Sorbonne University, LIB, Paris F-75006, France
| | - Giuseppe Spinelli
- Centre pour l'Acquisition et le Traitement des Images, (CATI platform), France; Laboratoire d'Imagerie Biomédicale, CNRS, INSERM, Sorbonne University, LIB, Paris F-75006, France
| | - Michel J Grothe
- German Center for Neurodegenerative Diseases (DZNE), Greifswald, Rostock, Germany
| | - Marie-Claude Potier
- Institut du Cerveau et de la Moelle épinière, ICM, INSERM U1127, CNRS UMR 7225, Sorbonne Université, Paris F- 75013, France
| | - Fabrizio Stocchi
- IRCCS San Raffaele, Rome, Italy; Telematic University, San Raffaele, Rome, Italy
| | | | - Marie-Odile Habert
- Centre pour l'Acquisition et le Traitement des Images, (CATI platform), France; Laboratoire d'Imagerie Biomédicale, CNRS, INSERM, Sorbonne University, LIB, Paris F-75006, France; AP-HP, Pitié-Salpêtrière Hospital, Department of Nuclear Medicine, Paris F-75013, France
| | - Bruno Dubois
- Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'hôpital, Paris F-75013, France; Institut du Cerveau et de la Moelle épinière, ICM, INSERM U1127, CNRS UMR 7225, Sorbonne Université, Paris F- 75013, France
| | - Claudio Babiloni
- Department of Physiology and Pharmacology "Erspamer", Sapienza University of Rome, Rome, Italy; San Raffaele Cassino, Cassino, FR, Italy.
| |
Collapse
|
19
|
Bouteloup V, Pellegrin I, Dubois B, Chene G, Planche V, Dufouil C. Explaining the Variability of Alzheimer Disease Fluid Biomarker Concentrations in Memory Clinic Patients Without Dementia. Neurology 2024; 102:e209219. [PMID: 38527237 PMCID: PMC11175632 DOI: 10.1212/wnl.0000000000209219] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 01/02/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Patients' comorbidities can affect Alzheimer disease (AD) blood biomarker concentrations. Because a limited number of factors have been explored to date, our aim was to assess the proportion of the variance in fluid biomarker levels explained by the clinical features of AD and by a large number of non-AD-related factors. METHODS MEMENTO enrolled 2,323 individuals with cognitive complaints or mild cognitive impairment in 26 French memory clinics. Baseline evaluation included clinical and neuropsychological assessments, brain MRI, amyloid-PET, CSF (optional), and blood sampling. Blood biomarker levels were determined using the Simoa-HDX analyzer. We performed linear regression analysis of the clinical features of AD (cognition, AD genetic risk score, and brain atrophy) to model biomarker concentrations. Next, we added covariates among routine biological tests, inflammatory markers, demographic and behavioral determinants, treatments, comorbidities, and preanalytical sample handling in final models using both stepwise selection processes and least absolute shrinkage and selection operator (LASSO). RESULTS In total, 2,257 participants were included in the analysis (median age 71.7, 61.8% women, 55.2% with high educational levels). For blood biomarkers, the proportion of variance explained by clinical features of AD was 13.7% for neurofilaments (NfL), 11.4% for p181-tau, 3.0% for Aβ-42/40, and 1.4% for total-tau. In final models accounting for non-AD-related factors, the variance was mainly explained by age, routine biological tests, inflammatory markers, and preanalytical sample handling. In CSF, the proportion of variance explained by clinical features of AD was 24.8% for NfL, 22.3% for Aβ-42/40, 19.8% for total-tau, and 17.2% for p181-tau. In contrast to blood biomarkers, the largest proportion of variance was explained by cognition after adjustment for covariates. The covariates that explained the largest proportion of variance were also the most frequently selected with LASSO. The performance of blood biomarkers for predicting A+ and T+ status (PET or CSF) remained unchanged after controlling for drivers of variance. DISCUSSION This comprehensive analysis demonstrated that the variance in AD blood biomarker concentrations was mainly explained by age, with minor contributions from cognition, brain atrophy, and genetics, conversely to CSF measures. These results challenge the use of blood biomarkers as isolated stand-alone biomarkers for AD.
Collapse
Affiliation(s)
- Vincent Bouteloup
- From the Univ. Bordeaux (V.B., G.C., C.D.), Inserm, Bordeaux Population Health, UMR1219, Bordeaux; CIC 1401 EC (V.B., G.C., C.D.), Pôle Santé Publique, CHU de Bordeaux; Laboratory of Immunology and Immunogenetics (I.P.), Resources Biological Center (CRB), CHU Bordeaux; Univ. Bordeaux (I.P.), CNRS, ImmunoConcEpT, UMR 5164, Bordeaux; Alzheimer Research Center IM2A (B.D.), Salpêtrière Hospital, AP-HP, Sorbonne University, Paris; Univ. Bordeaux (V.P.), CNRS, Institut des Maladies Neuroégénératives, UMR 5293, Bordeaux; Pôle de Neurosciences Cliniques (V.P.), Centre Mémoire de Ressources et de Recherche, CHU Bordeaux, France
| | - Isabelle Pellegrin
- From the Univ. Bordeaux (V.B., G.C., C.D.), Inserm, Bordeaux Population Health, UMR1219, Bordeaux; CIC 1401 EC (V.B., G.C., C.D.), Pôle Santé Publique, CHU de Bordeaux; Laboratory of Immunology and Immunogenetics (I.P.), Resources Biological Center (CRB), CHU Bordeaux; Univ. Bordeaux (I.P.), CNRS, ImmunoConcEpT, UMR 5164, Bordeaux; Alzheimer Research Center IM2A (B.D.), Salpêtrière Hospital, AP-HP, Sorbonne University, Paris; Univ. Bordeaux (V.P.), CNRS, Institut des Maladies Neuroégénératives, UMR 5293, Bordeaux; Pôle de Neurosciences Cliniques (V.P.), Centre Mémoire de Ressources et de Recherche, CHU Bordeaux, France
| | - Bruno Dubois
- From the Univ. Bordeaux (V.B., G.C., C.D.), Inserm, Bordeaux Population Health, UMR1219, Bordeaux; CIC 1401 EC (V.B., G.C., C.D.), Pôle Santé Publique, CHU de Bordeaux; Laboratory of Immunology and Immunogenetics (I.P.), Resources Biological Center (CRB), CHU Bordeaux; Univ. Bordeaux (I.P.), CNRS, ImmunoConcEpT, UMR 5164, Bordeaux; Alzheimer Research Center IM2A (B.D.), Salpêtrière Hospital, AP-HP, Sorbonne University, Paris; Univ. Bordeaux (V.P.), CNRS, Institut des Maladies Neuroégénératives, UMR 5293, Bordeaux; Pôle de Neurosciences Cliniques (V.P.), Centre Mémoire de Ressources et de Recherche, CHU Bordeaux, France
| | - Genevieve Chene
- From the Univ. Bordeaux (V.B., G.C., C.D.), Inserm, Bordeaux Population Health, UMR1219, Bordeaux; CIC 1401 EC (V.B., G.C., C.D.), Pôle Santé Publique, CHU de Bordeaux; Laboratory of Immunology and Immunogenetics (I.P.), Resources Biological Center (CRB), CHU Bordeaux; Univ. Bordeaux (I.P.), CNRS, ImmunoConcEpT, UMR 5164, Bordeaux; Alzheimer Research Center IM2A (B.D.), Salpêtrière Hospital, AP-HP, Sorbonne University, Paris; Univ. Bordeaux (V.P.), CNRS, Institut des Maladies Neuroégénératives, UMR 5293, Bordeaux; Pôle de Neurosciences Cliniques (V.P.), Centre Mémoire de Ressources et de Recherche, CHU Bordeaux, France
| | - Vincent Planche
- From the Univ. Bordeaux (V.B., G.C., C.D.), Inserm, Bordeaux Population Health, UMR1219, Bordeaux; CIC 1401 EC (V.B., G.C., C.D.), Pôle Santé Publique, CHU de Bordeaux; Laboratory of Immunology and Immunogenetics (I.P.), Resources Biological Center (CRB), CHU Bordeaux; Univ. Bordeaux (I.P.), CNRS, ImmunoConcEpT, UMR 5164, Bordeaux; Alzheimer Research Center IM2A (B.D.), Salpêtrière Hospital, AP-HP, Sorbonne University, Paris; Univ. Bordeaux (V.P.), CNRS, Institut des Maladies Neuroégénératives, UMR 5293, Bordeaux; Pôle de Neurosciences Cliniques (V.P.), Centre Mémoire de Ressources et de Recherche, CHU Bordeaux, France
| | - Carole Dufouil
- From the Univ. Bordeaux (V.B., G.C., C.D.), Inserm, Bordeaux Population Health, UMR1219, Bordeaux; CIC 1401 EC (V.B., G.C., C.D.), Pôle Santé Publique, CHU de Bordeaux; Laboratory of Immunology and Immunogenetics (I.P.), Resources Biological Center (CRB), CHU Bordeaux; Univ. Bordeaux (I.P.), CNRS, ImmunoConcEpT, UMR 5164, Bordeaux; Alzheimer Research Center IM2A (B.D.), Salpêtrière Hospital, AP-HP, Sorbonne University, Paris; Univ. Bordeaux (V.P.), CNRS, Institut des Maladies Neuroégénératives, UMR 5293, Bordeaux; Pôle de Neurosciences Cliniques (V.P.), Centre Mémoire de Ressources et de Recherche, CHU Bordeaux, France
| |
Collapse
|
20
|
Ferrari-Souza JP, Brum WS, Hauschild LA, Da Ros LU, Ferreira PCL, Bellaver B, Leffa DT, Bieger A, Tissot C, Lussier FZ, De Bastiani MA, Povala G, Benedet AL, Therriault J, Wang YT, Ashton NJ, Zetterberg H, Blennow K, Martins SO, Souza DO, Rosa-Neto P, Karikari TK, Pascoal TA, Zimmer ER. Vascular risk burden is a key player in the early progression of Alzheimer's disease. Neurobiol Aging 2024; 136:88-98. [PMID: 38335912 PMCID: PMC11416150 DOI: 10.1016/j.neurobiolaging.2023.12.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 12/21/2023] [Accepted: 12/29/2023] [Indexed: 02/12/2024]
Abstract
Understanding whether vascular risk factors (VRFs) synergistically potentiate Alzheimer's disease (AD) progression is important in the context of emerging treatments for preclinical AD. In a group of 503 cognitively unimpaired individuals, we tested whether VRF burden interacts with AD pathophysiology to accelerate neurodegeneration and cognitive decline. Baseline VRF burden was calculated considering medical data and AD pathophysiology was assessed based on cerebrospinal fluid (CSF) amyloid-β1-42 (Aβ1-42) and tau phosphorylated at threonine 181 (p-tau181). Neurodegeneration was assessed with plasma neurofilament light (NfL) and global cognition with the modified version of the Preclinical Alzheimer's Cognitive Composite. The mean (SD) age of participants was 72.9 (6.1) years, and 220 (43.7%) were men. Linear mixed-effects models revealed that an elevated VRF burden synergistically interacted with AD pathophysiology to drive longitudinal plasma NfL increase and cognitive decline. Additionally, VRF burden was not associated with CSF Aβ1-42 or p-tau181 changes over time. Our results suggest that VRF burden and AD pathophysiology are independent processes; however, they synergistically lead to neurodegeneration and cognitive deterioration. In preclinical stages, the combination of therapies targeting VRFs and AD pathophysiology might potentiate treatment outcomes.
Collapse
Affiliation(s)
- João Pedro Ferrari-Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wagner S Brum
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lucas A Hauschild
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Lucas U Da Ros
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Pâmela C L Ferreira
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruna Bellaver
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Douglas T Leffa
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andrei Bieger
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Cécile Tissot
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Firoza Z Lussier
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Marco Antônio De Bastiani
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Guilherme Povala
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andréa L Benedet
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden; Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden; Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway; Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China; UW Department of Medicine, School of Medicine and Public Health, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Sheila O Martins
- Department of Neurology, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Diogo O Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Thomas K Karikari
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Tharick A Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Graduate Program in Biological Sciences: Pharmacology and Therapeuctis, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
21
|
Ku YT, Chiu PY, Hua MS, Liao YC, Chang HT. Development of a Revised Version of Subjective Memory Complaints Questionnaire in Taiwan: A Preliminary Study. Arch Clin Neuropsychol 2024; 39:78-91. [PMID: 37337458 DOI: 10.1093/arclin/acad045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2023] [Indexed: 06/21/2023] Open
Abstract
OBJECTIVES Research has suggested that subjective memory complaints (SMC) are predictive of cognitive decline in cases of Alzheimer's disease; however, multidimensional characteristics of SMC make it difficult to formulate causal links. The Subjective Memory Complaints Questionnaire (SMCQ) has proven effective in capturing the nature of SMC. In this study, we developed a revised version of SMCQ (SMCQ-R) with corresponding normative data for application in Taiwan. METHODS This study recruited 100 cognitively normal participants (> 45 years) stratified according to demographic characteristics. Assessments were performed to evaluate test-retest reliability, criterion-related validity, and construct validity of SMCQ-R. SMCQ-R scores of 20 matched patients with mild cognitive impairment (MCI) were also compared with those of normal participants to test construct validity. RESULTS Reliability of SMCQ-R was satisfactory (0.81-0.95). Factor analysis revealed a three-factor structure: everyday memory problems (EMP), recent severe memory problems (RSMP), and long-term memory problems (LTMP). EMP and RSMP scores were negatively associated with objective cognitive function (r = -.20 to .39). Depressive symptoms were positively associated with all factors (r = .23-.33). Age was positively associated with total (b = 0.09, p < .05) and EMP scores (b = 0.06, p < .01). MCI patients obtained higher scores (p < .05) on all subscales. SMCQ-R scores discriminated between normal and MCI individuals (area under the curve = 0.77). This study established a norm based on scores adjusted to control for effects of age. CONCLUSIONS SMCQ-R has sound psychometric properties and could potentially be used as a tool to assess SMC in clinical settings.
Collapse
Affiliation(s)
- Yi-Ting Ku
- Department of Psychology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan
| | - Pai-Yi Chiu
- Department of Neurology, Show Chwan Memorial Hospital, Changhua City, Taiwan
- Department of Applied Mathematics, College of Science, Tunghai University, Taichung, Taiwan
| | - Mau-Sun Hua
- Department of Psychology, College of Science, National Taiwan University, Taipei, Taiwan
| | - Yu-Chi Liao
- Department of Psychology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan
- Clinical Psychology Center, Asia University, Taichung, Taiwan
| | - Hsin-Te Chang
- Department of Psychology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan
- Department of Psychology, College of Science, Chung Yuan Christian University, Taoyuan, Taiwan
- Research Assistance Center, Show Chwan Memorial Hospital, Changhua City, Taiwan
| |
Collapse
|
22
|
Rapos Pereira F, George N, Dalla Barba G, Dubois B, La Corte V. The Memory Binding Test Detects Early Subtle Episodic Memory Decline in Preclinical Alzheimer's Disease: A Longitudinal Study. J Alzheimers Dis 2024; 98:465-479. [PMID: 38393903 DOI: 10.3233/jad-230921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Background The asymptomatic at-risk phase might be the optimal time-window to establish clinically meaningful endpoints in Alzheimer's disease (AD). Objective We investigated whether, compared with the Free and Cued Selective Reminding Test (FCSRT), the Memory Binding Test (MBT) can anticipate the diagnosis of emergent subtle episodic memory (EM) deficits to an at-risk phase. Methods Five-year longitudinal FCSRT and MBT scores from 45 individuals matched for age, education, and gender, were divided into 3 groups of 15 subjects: Aβ-/controls, Aβ+/stable, and Aβ+/progressors (preclinical-AD). The MBT adds an associative memory component (binding), particularly sensitive to subtle EM decline. Results In the MBT, EM decline started in the Aβ+/progressors (preclinical-AD) up to 4 years prior to diagnosis in delayed free recall (FR), followed by decline in binding-associated scores 1 year later. Conversely, in the FCSRT, EM-decline began later, up to 3 years prior to diagnosis, in the same group on both immediate and delayed versions of FR, while on total recall (TR) and intrusions decline started only 1 year prior to diagnosis. Conclusions The MBT seems more sensitive than the FCSRT for early EM-decline detection, regarding the year of diagnosis and the number of scores showing AD-linked EM deficits (associated with the AD-characteristic amnesic hippocampal syndrome). Considering the MBT as a detection tool of early subtle EM-decline in an asymptomatic at-risk phase, and the FCSRT as a classification tool of stages of EM-decline from a preclinical phase, these tests ought to potentially become complementary diagnostic tools that can foster therapies to delay cognitive decline. Clinical trial registration title: Electrophysiological markers of the progression to clinical Alzheimer disease in asymptomatic at-risk individuals: a longitudinal event-related potential study of episodic memory in the INSIGHT pre-AD cohort (acronym: ePARAD).
Collapse
Affiliation(s)
- Filipa Rapos Pereira
- Institut du Cerveau - Paris Brain Institute - ICM, INSERM, U 1127, CNRS, UMR 7225' APHP, CENIR, Centre MEG-EEG, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
- Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière University Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | - Nathalie George
- Institut du Cerveau - Paris Brain Institute - ICM, INSERM, U 1127, CNRS, UMR 7225' APHP, CENIR, Centre MEG-EEG, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | | | - Bruno Dubois
- Institut du Cerveau - Paris Brain Institute - ICM, INSERM, U 1127, CNRS, UMR 7225' APHP, CENIR, Centre MEG-EEG, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
- Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière University Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
- Department of Neurology, Centre of Excellence of Neurodegenerative Disease (CoEN), ICM, CIC Neurosciences, Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Valentina La Corte
- Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière University Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
- Laboratoire Mémoire Cerveau et Cognition (UR 7536), Institut de Psychologie, Université Paris Cité, Boulogne-Billancourt, France
- Institut Universitaire de France, Paris, France
| |
Collapse
|
23
|
Zhou R, Wang L, Chen L, Feng X, Zhou R, Xiang P, Wen J, Huang Y, Zhou H. Bone Marrow-Derived GCA + Immune Cells Drive Alzheimer's Disease Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303402. [PMID: 37949676 PMCID: PMC10754099 DOI: 10.1002/advs.202303402] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/17/2023] [Indexed: 11/12/2023]
Abstract
Alzheimer's disease (AD) is an age-related degenerative disease of the central nervous system (CNS), whereas the role of bone marrow immune cells in the pathogenesis of AD remains unclear. Here, the study reveals that compared to matched healthy individuals, AD patients have higher circulating grancalcin (GCA) levels, which negatively correlate with cognitive function. Bone marrow-derived GCA+ immune cells, which secret abundant GCA and increase during aging, preferentially invaded the hippocampus and cortex of AD mouse model in a C-C Motif Chemokine Receptor 10 (CCR10)-dependent manner. Transplanting GCA+ immune cells or direct stereotaxic injection of recombinant GCA protein intensified amyloid plaque load and aggravated cognitive and memory impairments. In contrast, genetic ablation of GCA in the hematopoietic compartment improves cognitive and memory function. Mechanistically, GCA competitively binds to the low-density lipoprotein receptor-related protein 1 (LRP1) in microglia, thus inhibiting phagocytosis and clearance of Aβ and potentiating neuropathological changes. Importantly, GCA-neutralizing antibody treatment rejuvenated cognitive and memory function and constrained AD progression. Together, the study demonstrates a pathological role of GCA+ immune cells instigating cognitive and memory decline, suggesting that GCA+ immune cells can be a potential target for innovative therapeutic strategies in AD.
Collapse
Affiliation(s)
- Rui Zhou
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
| | - Liwen Wang
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
| | - Linyun Chen
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
| | - Xu Feng
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
| | - Ruoyu Zhou
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
| | - Peng Xiang
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
| | - Jie Wen
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
| | - Yan Huang
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalChangshaHunan410008China
| | - Haiyan Zhou
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalChangshaHunan410008China
| |
Collapse
|
24
|
Scheijbeler EP, de Haan W, Stam CJ, Twisk JWR, Gouw AA. Longitudinal resting-state EEG in amyloid-positive patients along the Alzheimer's disease continuum: considerations for clinical trials. Alzheimers Res Ther 2023; 15:182. [PMID: 37858173 PMCID: PMC10585755 DOI: 10.1186/s13195-023-01327-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 10/06/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND To enable successful inclusion of electroencephalography (EEG) outcome measures in Alzheimer's disease (AD) clinical trials, we retrospectively mapped the progression of resting-state EEG measures over time in amyloid-positive patients with mild cognitive impairment (MCI) or dementia due to AD. METHODS Resting-state 21-channel EEG was recorded in 148 amyloid-positive AD patients (MCI, n = 88; dementia due to AD, n = 60). Two or more EEG recordings were available for all subjects. We computed whole-brain and regional relative power (i.e., theta (4-8 Hz), alpha1 (8-10 Hz), alpha2 (10-13 Hz), beta (13-30 Hz)), peak frequency, signal variability (i.e., theta permutation entropy), and functional connectivity values (i.e., alpha and beta corrected amplitude envelope correlation, theta phase lag index, weighted symbolic mutual information, inverted joint permutation entropy). Whole-group linear mixed effects models were used to model the development of EEG measures over time. Group-wise analysis was performed to investigate potential differences in change trajectories between the MCI and dementia subgroups. Finally, we estimated the minimum sample size required to detect different treatment effects (i.e., 50% less deterioration, stabilization, or 50% improvement) on the development of EEG measures over time, in hypothetical clinical trials of 1- or 2-year duration. RESULTS Whole-group analysis revealed significant regional and global oscillatory slowing over time (i.e., increased relative theta power, decreased beta power), with strongest effects for temporal and parieto-occipital regions. Disease severity at baseline influenced the EEG measures' rates of change, with fastest deterioration reported in MCI patients. Only AD dementia patients displayed a significant decrease of the parieto-occipital peak frequency and theta signal variability over time. We estimate that 2-year trials, focusing on amyloid-positive MCI patients, require 36 subjects per arm (2 arms, 1:1 randomization, 80% power) to detect a stabilizing treatment effect on temporal relative theta power. CONCLUSIONS Resting-state EEG measures could facilitate early detection of treatment effects on neuronal function in AD patients. Their sensitivity depends on the region-of-interest and disease severity of the study population. Conventional spectral measures, particularly recorded from temporal regions, present sensitive AD treatment monitoring markers.
Collapse
Affiliation(s)
- Elliz P Scheijbeler
- Clinical Neurophysiology and MEG Center, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands.
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, Netherlands.
| | - Willem de Haan
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, Netherlands
| | - Cornelis J Stam
- Clinical Neurophysiology and MEG Center, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands
| | - Jos W R Twisk
- Department of Epidemiology and Biostatistics, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, Netherlands
| | - Alida A Gouw
- Clinical Neurophysiology and MEG Center, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, Netherlands
| |
Collapse
|
25
|
Giacomucci G, Mazzeo S, Crucitti C, Ingannato A, Bagnoli S, Padiglioni S, Galdo G, Emiliani F, Frigerio D, Moschini V, Morinelli C, Sorbi S, Bessi V, Nacmias B. Plasma p-tau181 as a promising non-invasive biomarker of Alzheimer's Disease pathology in Subjective Cognitive Decline and Mild Cognitive Impairment. J Neurol Sci 2023; 453:120805. [PMID: 37716237 DOI: 10.1016/j.jns.2023.120805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/21/2023] [Accepted: 09/09/2023] [Indexed: 09/18/2023]
Abstract
INTRODUCTION The aim of this study is to investigate the role of plasma phosphorylated tau (p-tau) 181 as a potential biomarker for Alzheimer's Disease (AD) pathology in the early stages of the disease, as a valuable marker for tauopathy. MATERIALS AND METHODS Thirty-three Subjective Cognitive Decline (SCD), 32 Mild Cognitive Impairment (MCI) and 14 AD demented (AD-d) patients underwent plasma p-tau181 analysis with SiMoA assay. Twenty-six SCD, 32 MCI and 14 AD-d patients also underwent CSF biomarkers analysis (Aβ1-42, Aβ1-42/1-40, p-tau, t-tau) and were classified as carriers of AD pathology (AP+) when A+ was associated with T+ (regardless of N), or non-carriers (AP-) when they were A- (regardless of T and N), or A+/T-/N-, or A+/T-/N+ according to the A/T(N) system. RESULTS Plasma p-tau181 levels were higher in SCD AP+ than in SCD AP- (2.85 ± 0.53 vs 1.73 ± 0.64, p < 0.001), and in MCI AP+ than in MCI AP- (4.03 ± 1.07 vs 2.04 ± 0.87, p < 0.001). In a multivariate linear regression analysis, AP status was the only variable that influenced plasma p-tau181 (B = 1.670 [95% CI 1.097:2.244], p < 0.001). Plasma p-tau181 was highly accurate for discriminating between AP+ and AP- patients (AUC = 0.910). We identified a cut-off level of 2.69 pg/mL to distinguish between AP+ and AP- (sensibility 0.86, specificity 0.82, PPV 75.00% NPV 90.32%). CONCLUSIONS Plasma p-tau181 levels were influenced by the presence of underlying AD pathology, independently from the cognitive status and were highly accurate in differentiating SCD-MCI patients who were carriers of AD pathology from non-carriers. Plasma p-tau181 might be a promising non-invasive biomarker of AD pathology at a very early stage.
Collapse
Affiliation(s)
- Giulia Giacomucci
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Salvatore Mazzeo
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Chiara Crucitti
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Assunta Ingannato
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Silvia Bagnoli
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Sonia Padiglioni
- Regional Referral Centre for Relational Criticalities - Tuscany Region, Italy; Research and Innovation Centre for Dementia-CRIDEM, AOU Careggi, Italy
| | - Giulia Galdo
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Filippo Emiliani
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Daniele Frigerio
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Valentina Moschini
- SOD Neurologia I, Dipartimento Neuromuscolo-Scheletrico e degli Organi di Senso, AOU Careggi, Florence, Italy
| | - Carmen Morinelli
- SOD Neurologia I, Dipartimento Neuromuscolo-Scheletrico e degli Organi di Senso, AOU Careggi, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Valentina Bessi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy.
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| |
Collapse
|
26
|
Gouilly D, Rafiq M, Nogueira L, Salabert AS, Payoux P, Péran P, Pariente J. Beyond the amyloid cascade: An update of Alzheimer's disease pathophysiology. Rev Neurol (Paris) 2023; 179:812-830. [PMID: 36906457 DOI: 10.1016/j.neurol.2022.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 10/02/2022] [Accepted: 12/02/2022] [Indexed: 03/13/2023]
Abstract
Alzheimer's disease (AD) is a multi-etiology disease. The biological system of AD is associated with multidomain genetic, molecular, cellular, and network brain dysfunctions, interacting with central and peripheral immunity. These dysfunctions have been primarily conceptualized according to the assumption that amyloid deposition in the brain, whether from a stochastic or a genetic accident, is the upstream pathological change. However, the arborescence of AD pathological changes suggests that a single amyloid pathway might be too restrictive or inconsistent with a cascading effect. In this review, we discuss the recent human studies of late-onset AD pathophysiology in an attempt to establish a general updated view focusing on the early stages. Several factors highlight heterogenous multi-cellular pathological changes in AD, which seem to work in a self-amplifying manner with amyloid and tau pathologies. Neuroinflammation has an increasing importance as a major pathological driver, and perhaps as a convergent biological basis of aging, genetic, lifestyle and environmental risk factors.
Collapse
Affiliation(s)
- D Gouilly
- Toulouse Neuroimaging Center, Toulouse, France.
| | - M Rafiq
- Toulouse Neuroimaging Center, Toulouse, France; Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France
| | - L Nogueira
- Department of Cell Biology and Cytology, CHU Toulouse Purpan, France
| | - A-S Salabert
- Toulouse Neuroimaging Center, Toulouse, France; Department of Nuclear Medicine, CHU Toulouse Purpan, France
| | - P Payoux
- Toulouse Neuroimaging Center, Toulouse, France; Department of Nuclear Medicine, CHU Toulouse Purpan, France; Center of Clinical Investigation, CHU Toulouse Purpan (CIC1436), France
| | - P Péran
- Toulouse Neuroimaging Center, Toulouse, France
| | - J Pariente
- Toulouse Neuroimaging Center, Toulouse, France; Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France; Center of Clinical Investigation, CHU Toulouse Purpan (CIC1436), France
| |
Collapse
|
27
|
Chu KT, Lei WC, Wu MH, Fuh JL, Wang SJ, French IT, Chang WS, Chang CF, Huang NE, Liang WK, Juan CH. A holo-spectral EEG analysis provides an early detection of cognitive decline and predicts the progression to Alzheimer's disease. Front Aging Neurosci 2023; 15:1195424. [PMID: 37674782 PMCID: PMC10477374 DOI: 10.3389/fnagi.2023.1195424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/25/2023] [Indexed: 09/08/2023] Open
Abstract
Aims Our aim was to differentiate patients with mild cognitive impairment (MCI) and Alzheimer's disease (AD) from cognitively normal (CN) individuals and predict the progression from MCI to AD within a 3-year longitudinal follow-up. A newly developed Holo-Hilbert Spectral Analysis (HHSA) was applied to resting state EEG (rsEEG), and features were extracted and subjected to machine learning algorithms. Methods A total of 205 participants were recruited from three hospitals, with CN (n = 51, MMSE > 26), MCI (n = 42, CDR = 0.5, MMSE ≥ 25), AD1 (n = 61, CDR = 1, MMSE < 25), AD2 (n = 35, CDR = 2, MMSE < 16), and AD3 (n = 16, CDR = 3, MMSE < 16). rsEEG was also acquired from all subjects. Seventy-two MCI patients (CDR = 0.5) were longitudinally followed up with two rsEEG recordings within 3 years and further subdivided into an MCI-stable group (MCI-S, n = 36) and an MCI-converted group (MCI-C, n = 36). The HHSA was then applied to the rsEEG data, and features were extracted and subjected to machine-learning algorithms. Results (a) At the group level analysis, the HHSA contrast of MCI and different stages of AD showed augmented amplitude modulation (AM) power of lower-frequency oscillations (LFO; delta and theta bands) with attenuated AM power of higher-frequency oscillations (HFO; beta and gamma bands) compared with cognitively normal elderly controls. The alpha frequency oscillation showed augmented AM power across MCI to AD1 with a reverse trend at AD2. (b) At the individual level of cross-sectional analysis, implementation of machine learning algorithms discriminated between groups with good sensitivity (Sen) and specificity (Spec) as follows: CN elderly vs. MCI: 0.82 (Sen)/0.80 (Spec), CN vs. AD1: 0.94 (Sen)/0.80 (Spec), CN vs. AD2: 0.93 (Sen)/0.90 (Spec), and CN vs. AD3: 0.75 (Sen)/1.00 (Spec). (c) In the longitudinal MCI follow-up, the initial contrasted HHSA between MCI-S and MCI-C groups showed significantly attenuated AM power of alpha and beta band oscillations. (d) At the individual level analysis of longitudinal MCI groups, deploying machine learning algorithms with the best seven features resulted in a sensitivity of 0.9 by the support vector machine (SVM) classifier, with a specificity of 0.8 yielded by the decision tree classifier. Conclusion Integrating HHSA into EEG signals and machine learning algorithms can differentiate between CN and MCI as well as also predict AD progression at the MCI stage.
Collapse
Affiliation(s)
- Kwo-Ta Chu
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
- Yang-Ming Hospital, Taoyuan, Taiwan
| | - Weng-Chi Lei
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
- Cognitive Intelligence and Precision Healthcare Center, National Central University, Taoyuan, Taiwan
| | - Ming-Hsiu Wu
- Division of Neurology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
- Department of Long-Term Care and Health Promotion, Min-Hwei Junior College of Health Care Management, Tainan, Taiwan
| | - Jong-Ling Fuh
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shuu-Jiun Wang
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Isobel T. French
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Central University and Academia Sinica, Taipei, Taiwan
| | - Wen-Sheng Chang
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
| | - Chi-Fu Chang
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
| | - Norden E. Huang
- Cognitive Intelligence and Precision Healthcare Center, National Central University, Taoyuan, Taiwan
- Key Laboratory of Data Analysis and Applications, First Institute of Oceanography, SOA, Qingdao, China
| | - Wei-Kuang Liang
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
- Cognitive Intelligence and Precision Healthcare Center, National Central University, Taoyuan, Taiwan
| | - Chi-Hung Juan
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
- Cognitive Intelligence and Precision Healthcare Center, National Central University, Taoyuan, Taiwan
- Department of Psychology, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
28
|
Devi G. A how-to guide for a precision medicine approach to the diagnosis and treatment of Alzheimer's disease. Front Aging Neurosci 2023; 15:1213968. [PMID: 37662550 PMCID: PMC10469885 DOI: 10.3389/fnagi.2023.1213968] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/24/2023] [Indexed: 09/05/2023] Open
Abstract
Article purpose The clinical approach to Alzheimer's disease (AD) is challenging, particularly in high-functioning individuals. Accurate diagnosis is crucial, especially given the significant side effects, including brain hemorrhage, of newer monoclonal antibodies approved for treating earlier stages of Alzheimer's. Although early treatment is more effective, early diagnosis is also more difficult. Several clinical mimickers of AD exist either separately, or in conjunction with AD pathology, adding to the diagnostic complexity. To illustrate the clinical decision-making process, this study includes de-identified cases and reviews of the underlying etiology and pathology of Alzheimer's and available therapies to exemplify diagnostic and treatment subtleties. Problem The clinical presentation of Alzheimer's is complex and varied. Multiple other primary brain pathologies present with clinical phenotypes that can be difficult to distinguish from AD. Furthermore, Alzheimer's rarely exists in isolation, as almost all patients also show evidence of other primary brain pathologies, including Lewy body disease and argyrophilic grain disease. The phenotype and progression of AD can vary based on the brain regions affected by pathology, the coexistence and severity of other brain pathologies, the presence and severity of systemic comorbidities such as cardiac disease, the common co-occurrence with psychiatric diagnoses, and genetic risk factors. Additionally, symptoms and progression are influenced by an individual's brain reserve and cognitive reserve, as well as the timing of the diagnosis, which depends on the demographics of both the patient and the diagnosing physician, as well as the availability of biomarkers. Methods The optimal clinical and biomarker strategy for accurately diagnosing AD, common neuropathologic co-morbidities and mimickers, and available medication and non-medication-based treatments are discussed. Real-life examples of cognitive loss illustrate the diagnostic and treatment decision-making process as well as illustrative treatment responses. Implications AD is best considered a syndromic disorder, influenced by a multitude of patient and environmental characteristics. Additionally, AD existing alone is a unicorn, as there are nearly always coexisting other brain pathologies. Accurate diagnosis with biomarkers is essential. Treatment response is affected by the variables involved, and the effective treatment of Alzheimer's disease, as well as its prevention, requires an individualized, precision medicine strategy.
Collapse
Affiliation(s)
- Gayatri Devi
- Neurology and Psychiatry, Zucker School of Medicine, Hempstead, NY, United States
- Neurology and Psychiatry, Lenox Hill Hospital, New York City, NY, United States
- Park Avenue Neurology, New York City, NY, United States
| |
Collapse
|
29
|
Ulbl J, Rakusa M. The Importance of Subjective Cognitive Decline Recognition and the Potential of Molecular and Neurophysiological Biomarkers-A Systematic Review. Int J Mol Sci 2023; 24:10158. [PMID: 37373304 DOI: 10.3390/ijms241210158] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/01/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Subjective cognitive decline (SCD) and mild cognitive impairment (MCI) are early stages of Alzheimer's disease (AD). Neurophysiological markers such as electroencephalography (EEG) and event-related potential (ERP) are emerging as alternatives to traditional molecular and imaging markers. This paper aimed to review the literature on EEG and ERP markers in individuals with SCD. We analysed 30 studies that met our criteria, with 17 focusing on resting-state or cognitive task EEG, 11 on ERPs, and two on both EEG and ERP parameters. Typical spectral changes were indicative of EEG rhythm slowing and were associated with faster clinical progression, lower education levels, and abnormal cerebrospinal fluid biomarkers profiles. Some studies found no difference in ERP components between SCD subjects, controls, or MCI, while others reported lower amplitudes in the SCD group compared to controls. Further research is needed to explore the prognostic value of EEG and ERP in relation to molecular markers in individuals with SCD.
Collapse
Affiliation(s)
- Janina Ulbl
- Division of Neurology, University Medical Centre Maribor, 2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Martin Rakusa
- Division of Neurology, University Medical Centre Maribor, 2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| |
Collapse
|
30
|
Lesoil C, Bombois S, Guinebretiere O, Houot M, Bahrami M, Levy M, Genthon R, Bozon F, Jean-Marie H, Epelbaum S, Foulon P, Villain N, Dubois B. Validation study of "Santé-Cerveau", a digital tool for early cognitive changes identification. Alzheimers Res Ther 2023; 15:70. [PMID: 37013590 PMCID: PMC10068729 DOI: 10.1186/s13195-023-01204-x] [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: 12/05/2022] [Accepted: 03/11/2023] [Indexed: 04/05/2023]
Abstract
BACKGROUND There is a need for a reliable, easy-to-use, widely available, and validated tool for timely cognitive impairment identification. We created a computerized cognitive screening tool (Santé-Cerveau digital tool (SCD-T)) including validated questionnaires and the following neuropsychological tests: 5 Word Test (5-WT) for episodic memory, Trail Making Test (TMT) for executive functions, and a number coding test (NCT) adapted from the Digit Symbol Substitution Test for global intellectual efficiency. This study aimed to evaluate the performance of SCD-T to identify cognitive deficit and to determine its usability. METHODS Three groups were constituted including 65 elderly Controls, 64 patients with neurodegenerative diseases (NDG): 50 AD and 14 non-AD, and 20 post-COVID-19 patients. The minimum MMSE score for inclusion was 20. Association between computerized SCD-T cognitive tests and their standard equivalent was assessed using Pearson's correlation coefficients. Two algorithms (a simple clinician-guided algorithm involving the 5-WT and the NCT; and a machine learning classifier based on 8 scores from the SCD-T tests extracted from a multiple logistic regression model, and data from the SCD-T questionnaires) were evaluated. The acceptability of SCD-T was investigated through a questionnaire and scale. RESULTS AD and non-AD participants were older (mean ± standard deviation (SD): 72.61 ± 6.79 vs 69.91 ± 4.86 years old, p = 0.011) and had a lower MMSE score (Mean difference estimate ± standard error: 1.74 ± 0.14, p < 0.001) than Controls; post-COVID-19 patients were younger than Controls (mean ± SD: 45.07 ± 11.36 years old, p < 0.001). All the computerized SCD-T cognitive tests were significantly associated with their reference version. In the pooled Controls and NDG group, the correlation coefficient was 0.84 for verbal memory, -0.60 for executive functions, and 0.72 for global intellectual efficiency. The clinician-guided algorithm demonstrated 94.4% ± 3.8% sensitivity and 80.5% ± 8.7% specificity, and the machine learning classifier 96.8% ± 3.9% sensitivity and 90.7% ± 5.8% specificity. The acceptability of SCD-T was good to excellent. CONCLUSIONS We demonstrate the high accuracy of SCD-T in screening cognitive disorders and its good acceptance even in individuals with prodromal and mild dementia stages. SCD-T would be useful in primary care to faster refer subjects with significant cognitive impairment (and limit unnecessary referrals) to specialized consultation, improve the AD care pathway and the pre-screening in clinical trials.
Collapse
Affiliation(s)
- Constance Lesoil
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Paris, France
| | - Stéphanie Bombois
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Paris, France
- INSERM U1171 - Degenerative and Vascular Cognitive Disorders, University of Lille, Lille, France
| | - Octave Guinebretiere
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Paris, France
| | - Marion Houot
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Paris, France
- Centre of Excellence of Neurodegenerative Disease (CoEN), AP-HP, Pitié-Salpêtrière, Institut du Cerveau - Paris Brain Institute - ICM, Paris, France
| | - Mahsa Bahrami
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Paris, France
| | - Marcel Levy
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Paris, France
| | - Rémy Genthon
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Paris, France
| | - Frédérique Bozon
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Paris, France
| | | | - Stéphane Epelbaum
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Paris, France
- Centre of Excellence of Neurodegenerative Disease (CoEN), AP-HP, Pitié-Salpêtrière, Institut du Cerveau - Paris Brain Institute - ICM, Paris, France
| | | | - Nicolas Villain
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Paris, France
- Centre of Excellence of Neurodegenerative Disease (CoEN), AP-HP, Pitié-Salpêtrière, Institut du Cerveau - Paris Brain Institute - ICM, Paris, France
- Sorbonne Université, INSERM U1127, CNRS 7225, Paris, France
| | - Bruno Dubois
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Paris, France.
- Centre of Excellence of Neurodegenerative Disease (CoEN), AP-HP, Pitié-Salpêtrière, Institut du Cerveau - Paris Brain Institute - ICM, Paris, France.
- Sorbonne Université, INSERM U1127, CNRS 7225, Paris, France.
| |
Collapse
|
31
|
Liu KY, Thambisetty M, Howard R. How can secondary dementia prevention trials of Alzheimer's disease be clinically meaningful? Alzheimers Dement 2023; 19:1073-1085. [PMID: 36161763 PMCID: PMC10039957 DOI: 10.1002/alz.12788] [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: 04/21/2022] [Revised: 07/06/2022] [Accepted: 08/09/2022] [Indexed: 11/08/2022]
Abstract
After clinical trial failures in symptomatic Alzheimer's disease (AD), our field has moved to earlier intervention in cognitively normal individuals with biomarker evidence of AD. This offers potential for dementia prevention, but mainly low and variable rates of progression to AD dementia reduce the usefulness of trials' data in decision making by potential prescribers. With results from several Phase 3 secondary prevention studies anticipated within the next few years and the Food and Drug Administration's recent endorsement of amyloid beta as a surrogate outcome biomarker for AD clinical trials, it is time to question the clinical significance of changes in biomarkers, adequacy of current trial durations, and criteria for treatment success if cognitively unimpaired patients and their doctors are to meaningfully evaluate the potential value of new agents. We argue for a change of direction toward trial designs that can unambiguously inform clinical decision making about dementia risk and progression.
Collapse
Affiliation(s)
- Kathy Y. Liu
- Division of Psychiatry, University College London, London, UK
| | - Madhav Thambisetty
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, USA
| | - Robert Howard
- Division of Psychiatry, University College London, London, UK
| |
Collapse
|
32
|
Groechel RC, Tripodis Y, Alosco ML, Mez J, Qiu WQ, Mercier G, Goldstein L, Budson AE, Kowall N, Killiany RJ. Annualized changes in rate of amyloid deposition and neurodegeneration are greater in participants who become amyloid positive than those who remain amyloid negative. Neurobiol Aging 2023; 127:33-42. [PMID: 37043881 DOI: 10.1016/j.neurobiolaging.2023.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 03/01/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023]
Abstract
This study longitudinally examined participants in the Alzheimer's Disease Neuroimaging Initiative (ADNI) who underwent a conversion in amyloid-beta (Aβ) status in comparison to a group of ADNI participants who did not show a change in amyloid status over the same follow-up period. Participants included 136 ADNI dementia-free participants with 2 florbetapir positron emission tomography (PET) scans. Of these participants, 68 showed amyloid conversion as measured on florbetapir PET, and the other 68 did not. Amyloid converters and non-converters were chosen to have representative demographic data (age, education, sex, diagnostic status, and race). The amyloid converter group showed increased prevalence of APOE ε4 (p < 0.001), greater annualized percent volume loss in selected magnetic resonance imaging (MRI) regions (p < 0.05), lower cerebrospinal fluid Aβ1-42 (p < 0.001), and greater amyloid retention (as measured by standard uptake value ratios) on florbetapir PET scans (p < 0.001) in comparison to the non-converter group. These results provide compelling evidence that important neuropathological changes are occurring alongside amyloid conversion.
Collapse
|
33
|
Jessen F, Wolfsgruber S, Kleineindam L, Spottke A, Altenstein S, Bartels C, Berger M, Brosseron F, Daamen M, Dichgans M, Dobisch L, Ewers M, Fenski F, Fliessbach K, Freiesleben SD, Glanz W, Görß D, Gürsel S, Janowitz D, Kilimann I, Kobeleva X, Lohse A, Maier F, Metzger C, Munk M, Preis L, Sanzenbacher C, Spruth E, Rauchmann B, Vukovich R, Yakupov R, Weyrauch AS, Ziegler G, Schmid M, Laske C, Perneczky R, Schneider A, Wiltfang J, Teipel S, Bürger K, Priller J, Peters O, Ramirez A, Boecker H, Heneka MT, Wagner M, Düzel E. Subjective cognitive decline and stage 2 of Alzheimer disease in patients from memory centers. Alzheimers Dement 2023; 19:487-497. [PMID: 35451563 DOI: 10.1002/alz.12674] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/22/2022] [Accepted: 02/17/2022] [Indexed: 11/06/2022]
Abstract
INTRODUCTION It is uncertain whether subjective cognitive decline (SCD) in individuals who seek medical help serves the identification of the initial symptomatic stage 2 of the Alzheimer's disease (AD) continuum. METHODS Cross-sectional and longitudinal data from the multicenter, memory clinic-based DELCODE study. RESULTS The SCD group showed slightly worse cognition as well as more subtle functional and behavioral symptoms than the control group (CO). SCD-A+ cases (39.3% of all SCD) showed greater hippocampal atrophy, lower cognitive and functional performance, and more behavioral symptoms than CO-A+. Amyloid concentration in the CSF had a greater effect on longitudinal cognitive decline in SCD than in the CO group. DISCUSSION Our data suggests that SCD serves the identification of stage 2 of the AD continuum and that stage 2, operationalized as SCD-A+, is associated with subtle, but extended impact of AD pathology in terms of neurodegeneration, symptoms and clinical progression.
Collapse
Affiliation(s)
- Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, Germany
| | - Steffen Wolfsgruber
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn, Bonn, Germany
| | - Luca Kleineindam
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn, Bonn, Germany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurology, University of Bonn, Bonn, Germany
| | - Slawek Altenstein
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.,Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Bartels
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, Goettingen, Germany
| | - Moritz Berger
- Institute for Medical Biometry, University of Bonn, Bonn, Germany
| | | | - Marcel Daamen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Radiology, University of Bonn, Bonn, Germany
| | - Martin Dichgans
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany
| | - Laura Dobisch
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute for Cognitive Neurology and Dementia Research, University of Magdeburg, Magdeburg, Germany
| | - Michael Ewers
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany
| | - Friederike Fenski
- Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Klaus Fliessbach
- Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn, Bonn, Germany
| | | | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute for Cognitive Neurology and Dementia Research, University of Magdeburg, Magdeburg, Germany
| | - Doreen Görß
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany.,Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| | - Selim Gürsel
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University, München, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany.,Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| | - Xenia Kobeleva
- Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn, Bonn, Germany
| | - Andrea Lohse
- Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Franziska Maier
- Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, Germany
| | - Coraline Metzger
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute for Cognitive Neurology and Dementia Research, University of Magdeburg, Magdeburg, Germany
| | - Matthias Munk
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Lukas Preis
- Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Carolin Sanzenbacher
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Eike Spruth
- Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Boris Rauchmann
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University, München, Germany
| | - Ruth Vukovich
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, Goettingen, Germany
| | - Renat Yakupov
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute for Cognitive Neurology and Dementia Research, University of Magdeburg, Magdeburg, Germany
| | - Anne-Sophie Weyrauch
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn, Bonn, Germany
| | - Gabriel Ziegler
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute for Cognitive Neurology and Dementia Research, University of Magdeburg, Magdeburg, Germany
| | - Matthias Schmid
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Institute for Medical Biometry, University of Bonn, Bonn, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University, München, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn, Bonn, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, Goettingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany.,Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| | - Katharina Bürger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.,Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.,Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Alfredo Ramirez
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, Germany.,Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn, Bonn, Germany
| | - Henning Boecker
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Radiology, University of Bonn, Bonn, Germany
| | - Michael T Heneka
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn, Bonn, Germany
| | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn, Bonn, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute for Cognitive Neurology and Dementia Research, University of Magdeburg, Magdeburg, Germany
| |
Collapse
|
34
|
Macoir J, Tremblay P, Hudon C. The Use of Executive Fluency Tasks to Detect Cognitive Impairment in Individuals with Subjective Cognitive Decline. Behav Sci (Basel) 2022; 12:491. [PMID: 36546974 PMCID: PMC9774264 DOI: 10.3390/bs12120491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/10/2022] Open
Abstract
OBJECTIVE Although evidence has indicated that subjective cognitive decline (SCD) may be an early sign of Alzheimer's disease (AD), the objectification of cognitive impairment in SCD is challenging, mainly due to the lack of sensitivity in assessment tools. The present study investigated the potential contribution of two verbal fluency tasks with high executive processing loads to the identification of cognitive impairment in SCD. METHODS A total of 60 adults with SCD and 60 healthy controls (HCs) performed one free action (verb) fluency task and two fluency tasks with more executive processing load-an alternating fluency task and an orthographic constraint fluency task-and the results were compared. RESULT In the free action fluency task, the performance of the participants with SCD and the HCs was similar. However, HCs performed significantly better than SCD in the alternating fluency task, which required mental flexibility, and the orthographic constraint fluency task, which required inhibition. DISCUSSION The study findings suggest that verbal fluency tasks with high executive processing load could be useful in detecting cognitive deficits at the preclinical stage of AD. The inclusion of such tests in assessment batteries should be considered in order to improve the detection of subtle cognitive impairment in preclinical major neurocognitive disorder populations.
Collapse
Affiliation(s)
- Joël Macoir
- Faculté de Médecine, Département de Réadaptation, Université Laval, Québec, QC G1V 0A6, Canada
- Centre de Recherche CERVO—Brain Research Centre, Québec, QC G1J 2G3, Canada
| | - Pascale Tremblay
- Faculté de Médecine, Département de Réadaptation, Université Laval, Québec, QC G1V 0A6, Canada
- Centre de Recherche CERVO—Brain Research Centre, Québec, QC G1J 2G3, Canada
| | - Carol Hudon
- Centre de Recherche CERVO—Brain Research Centre, Québec, QC G1J 2G3, Canada
- Faculté des Sciences Sociales, École de Psychologie, Université Laval, Québec, QC G1V 0A6, Canada
- Centre de Recherche VITAM, Québec, QC G1J 2G1, Canada
| |
Collapse
|
35
|
Lazar AN, Hanbouch L, Boussicaut L, Fourmaux B, Daira P, Millan MJ, Bernoud-Hubac N, Potier MC. Lipid Dys-Homeostasis Contributes to APOE4-Associated AD Pathology. Cells 2022; 11:cells11223616. [PMID: 36429044 PMCID: PMC9688773 DOI: 10.3390/cells11223616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/21/2022] [Accepted: 11/09/2022] [Indexed: 11/18/2022] Open
Abstract
The association of the APOE4 (vs. APOE3) isoform with an increased risk of Alzheimer's disease (AD) is unequivocal, but the underlying mechanisms remain incompletely elucidated. A prevailing hypothesis incriminates the impaired ability of APOE4 to clear neurotoxic amyloid-β peptides (Aβ) from the brain as the main mechanism linking the apolipoprotein isoform to disease etiology. The APOE protein mediates lipid transport both within the brain and from the brain to the periphery, suggesting that lipids may be potential co-factors in APOE4-associated physiopathology. The present study reveals several changes in the pathways of lipid homeostasis in the brains of mice expressing the human APOE4 vs. APOE3 isoform. Carriers of APOE4 had altered cholesterol turnover, an imbalance in the ratio of specific classes of phospholipids, lower levels of phosphatidylethanolamines bearing polyunsaturated fatty acids and an overall elevation in levels of monounsaturated fatty acids. These modifications in lipid homeostasis were related to increased production of Aβ peptides as well as augmented levels of tau and phosphorylated tau in primary neuronal cultures. This suite of APOE4-associated anomalies in lipid homeostasis and neurotoxic protein levels may be related to the accrued risk for AD in APOE4 carriers and provides novel insights into potential strategies for therapeutic intervention.
Collapse
Affiliation(s)
- Adina-Nicoleta Lazar
- Univ Lyon, INSA Lyon, CNRS, LaMCoS, UMR5259, 69621 Villeurbanne, France
- Correspondence: (A.-N.L.); (M.-C.P.)
| | - Linda Hanbouch
- ICM Paris Brain Institute, CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière, 47 Bd de l’Hôpital, 75013 Paris, France
| | - Lydie Boussicaut
- ICM Paris Brain Institute, CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière, 47 Bd de l’Hôpital, 75013 Paris, France
| | - Baptiste Fourmaux
- Univ Lyon, INSA Lyon, CNRS, LaMCoS, UMR5259, 69621 Villeurbanne, France
| | - Patricia Daira
- Univ Lyon, INSA Lyon, CNRS, LaMCoS, UMR5259, 69621 Villeurbanne, France
| | - Mark J. Millan
- Institut De Recherche Servier IDRS, Neuroscience Inflammation Thérapeutic Area, 125 Chemin de Ronde, 78290 Croissy-sur-Seine, France
- Institute of Neuroscience and Psychology, College of Medical, Vet and life Sciences, Glasgow University, 68 Hillhead Street, Glasgow G12 8QB, Scotland, UK
| | | | - Marie-Claude Potier
- ICM Paris Brain Institute, CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière, 47 Bd de l’Hôpital, 75013 Paris, France
- Correspondence: (A.-N.L.); (M.-C.P.)
| |
Collapse
|
36
|
Alzheimer's Disease: Treatment Strategies and Their Limitations. Int J Mol Sci 2022; 23:ijms232213954. [PMID: 36430432 PMCID: PMC9697769 DOI: 10.3390/ijms232213954] [Citation(s) in RCA: 200] [Impact Index Per Article: 66.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is the most frequent case of neurodegenerative disease and is becoming a major public health problem all over the world. Many therapeutic strategies have been explored for several decades; however, there is still no curative treatment, and the priority remains prevention. In this review, we present an update on the clinical and physiological phase of the AD spectrum, modifiable and non-modifiable risk factors for AD treatment with a focus on prevention strategies, then research models used in AD, followed by a discussion of treatment limitations. The prevention methods can significantly slow AD evolution and are currently the best strategy possible before the advanced stages of the disease. Indeed, current drug treatments have only symptomatic effects, and disease-modifying treatments are not yet available. Drug delivery to the central nervous system remains a complex process and represents a challenge for developing therapeutic and preventive strategies. Studies are underway to test new techniques to facilitate the bioavailability of molecules to the brain. After a deep study of the literature, we find the use of soft nanoparticles, in particular nanoliposomes and exosomes, as an innovative approach for preventive and therapeutic strategies in reducing the risk of AD and solving problems of brain bioavailability. Studies show the promising role of nanoliposomes and exosomes as smart drug delivery systems able to penetrate the blood-brain barrier and target brain tissues. Finally, the different drug administration techniques for neurological disorders are discussed. One of the promising therapeutic methods is the intranasal administration strategy which should be used for preclinical and clinical studies of neurodegenerative diseases.
Collapse
|
37
|
Whelan R, Barbey FM, Cominetti MR, Gillan CM, Rosická AM. Developments in scalable strategies for detecting early markers of cognitive decline. Transl Psychiatry 2022; 12:473. [PMID: 36351888 PMCID: PMC9645320 DOI: 10.1038/s41398-022-02237-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 11/10/2022] Open
Abstract
Effective strategies for early detection of cognitive decline, if deployed on a large scale, would have individual and societal benefits. However, current detection methods are invasive or time-consuming and therefore not suitable for longitudinal monitoring of asymptomatic individuals. For example, biological markers of neuropathology associated with cognitive decline are typically collected via cerebral spinal fluid, cognitive functioning is evaluated from face-to-face assessments by experts and brain measures are obtained using expensive, non-portable equipment. Here, we describe scalable, repeatable, relatively non-invasive and comparatively inexpensive strategies for detecting the earliest markers of cognitive decline. These approaches are characterized by simple data collection protocols conducted in locations outside the laboratory: measurements are collected passively, by the participants themselves or by non-experts. The analysis of these data is, in contrast, often performed in a centralized location using sophisticated techniques. Recent developments allow neuropathology associated with potential cognitive decline to be accurately detected from peripheral blood samples. Advances in smartphone technology facilitate unobtrusive passive measurements of speech, fine motor movement and gait, that can be used to predict cognitive decline. Specific cognitive processes can be assayed using 'gamified' versions of standard laboratory cognitive tasks, which keep users engaged across multiple test sessions. High quality brain data can be regularly obtained, collected at-home by users themselves, using portable electroencephalography. Although these methods have great potential for addressing an important health challenge, there are barriers to be overcome. Technical obstacles include the need for standardization and interoperability across hardware and software. Societal challenges involve ensuring equity in access to new technologies, the cost of implementation and of any follow-up care, plus ethical issues.
Collapse
Affiliation(s)
- Robert Whelan
- School of Psychology, Trinity College Dublin, Dublin, Ireland.
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.
| | - Florentine M Barbey
- School of Psychology, Trinity College Dublin, Dublin, Ireland
- Cumulus Neuroscience Ltd, Dublin, Ireland
| | - Marcia R Cominetti
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
- Department of Gerontology, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Claire M Gillan
- School of Psychology, Trinity College Dublin, Dublin, Ireland
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Anna M Rosická
- School of Psychology, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
38
|
Baldeiras I, Silva-Spínola A, Lima M, Leitão MJ, Durães J, Vieira D, Tbuas-Pereira M, Cruz VT, Rocha R, Alves L, Machado Á, Milheiro M, Santiago B, Santana I. Alzheimer’s Disease Diagnosis Based on the Amyloid, Tau, and Neurodegeneration Scheme (ATN) in a Real-Life Multicenter Cohort of General Neurological Centers. J Alzheimers Dis 2022; 90:419-432. [DOI: 10.3233/jad-220587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: The ATN scheme was proposed as an unbiased biological characterization of the Alzheimer’s disease (AD) spectrum, grouping biomarkers into three categories: brain Amyloidosis-A, Tauopathy-T, Neurodegeneration-N. Although this scheme was mainly recommended for research, it is relevant for diagnosis. Objective: To evaluate the ATN scheme performance in real-life cohorts reflecting the inflow of patients with cognitive complaints and different underlying disorders in general neurological centers. Methods: We included patients (n = 1,128) from six centers with their core cerebrospinal fluid-AD biomarkers analyzed centrally. A was assessed through Aβ 42/Aβ 40, T through pTau-181, and N through tTau. Association between demographic features, clinical diagnosis at baseline/follow-up and ATN profiles was assessed. Results: The prevalence of ATN categories was: A-T-N-: 28.3% ; AD continuum (A + T-/+N-/+): 47.8% ; non-AD (A- plus T or/and N+): 23.9% . ATN profiles prevalence was strongly influenced by age, showing differences according to gender, APOE genotype, and cognitive status. At baseline, 74.6% of patients classified as AD fell in the AD continuum, decreasing to 47.4% in mild cognitive impairment and 42.3% in other neurodegenerative conditions. At follow-up, 41% of patients changed diagnosis, and 92% of patients that changed to AD were classified within the AD continuum. A + was the best individual marker for predicting a final AD diagnosis, and the combinations A + T+(irrespective of N) and A + T+N+had the highest overall accuracy (83%). Conclusion: The ATN scheme is useful to guide AD diagnosis real-life neurological centers settings. However, it shows a lack of accuracy for patients with other types of dementia. In such cases, the inclusion of other markers specific for non-AD proteinopathies could be an important aid to the differential diagnosis.
Collapse
Affiliation(s)
- Inês Baldeiras
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Anuschka Silva-Spínola
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Marisa Lima
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Center for Research in Neuropsychology and Cognitive Behavioral Intervention, Faculty of Psychology and Educational Sciences, University of Coimbra, Coimbra, Portugal
| | - Maria João Leitão
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - João Durães
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Daniela Vieira
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Miguel Tbuas-Pereira
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | | | - Raquel Rocha
- ULSM Unidade Local de Sáude de Matosinhos, Matosinhos, Portugal
| | - Luisa Alves
- Hospital Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Lisboa, Portugal
| | | | | | | | - Isabel Santana
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
39
|
Chen B, Fu Y, Song G, Zhong W, Guo J. Research trends and hotspots of exercise for Alzheimer’s disease: A bibliometric analysis. Front Aging Neurosci 2022; 14:984705. [PMID: 36158544 PMCID: PMC9490271 DOI: 10.3389/fnagi.2022.984705] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Alzheimer’s disease (AD) is a socially significant neurodegenerative disorder among the elderly worldwide. An increasing number of studies have revealed that as a non-pharmacological intervention, exercise can prevent and treat AD. However, information regarding the research status of this field remains minimal. Therefore, this study aimed to analyze trends and topics in exercise and AD research by using a bibliometric method. Methods We systematically searched the Web of Science Core Collection for published papers on exercise and AD. The retrieved data regarding institutions, journals, countries, authors, journal distribution, and keywords were analyzed using CiteSpace software. Meanwhile, the co-occurrence of keywords was constructed. Results A total of 1,104 papers were ultimately included in accordance with our specified inclusion criteria. The data showed that the number of published papers on exercise and AD is increasing each year, with papers published in 64 countries/regions and 396 academic journals. The Journal of Alzheimer’s Disease published the most papers (73 publications). Journals are concentrated in the fields of neuroscience and geriatrics gerontology. The University of Kansas and the United States are the major institution and country, respectively. The cited keywords show that oxidative stress, amyloid beta, and physical exercise are the research hotspots in recent years. After analysis, the neuroprotective effect of exercise was identified as the development trend in this field. Conclusions Based on a bibliometric analysis, the number of publications on exercise and AD has been increasing rapidly, especially in the past 10 years. “Amyloid beta,” “oxidative stress,” and “exercise program” trigger the most interest among researchers in this field. The study of exercise program and mechanism of exercise in AD is still the focus of future research.
Collapse
Affiliation(s)
- Binglin Chen
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Yujie Fu
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Ge Song
- Department of Rehabilitation Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiquan Zhong
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Jiabao Guo
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Jiabao Guo,
| |
Collapse
|
40
|
Spinelli G, Bakardjian H, Schwartz D, Potier MC, Habert MO, Levy M, Dubois B, George N. Theta Band-Power Shapes Amyloid-Driven Longitudinal EEG Changes in Elderly Subjective Memory Complainers At-Risk for Alzheimer's Disease. J Alzheimers Dis 2022; 90:69-84. [PMID: 36057818 DOI: 10.3233/jad-220204] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) includes progressive symptoms spread along a continuum of preclinical and clinical stages. Although numerous studies uncovered the neuro-cognitive changes of AD, very little is known on the natural history of brain lesions and modifications of brain networks in elderly cognitively-healthy memory complainers at risk of AD for carrying pathophysiological biomarkers (amyloidopathy and tauopathy). OBJECTIVE We analyzed resting-state electroencephalography (EEG) of 318 cognitively-healthy subjective memory complainers from the INSIGHT-preAD cohort at the time of their first visit (M0) and two-years later (M24). METHODS Using 18F-florbetapir PET-scanner, subjects were stratified between amyloid negative (A-; n = 230) and positive (A+; n = 88) groups. Differences between A+ and A-were estimated at source-level in each band-power of the EEG spectrum. RESULTS At M0, we found an increase of theta power in the mid-frontal cortex in A+ compared to A-. No significant association was found between mid-frontal theta and the individuals' cognitive performance. At M24, theta power increased in A+ relative to A-individuals in the posterior cingulate cortex and the pre-cuneus. Alpha band revealed a peculiar decremental trend in posterior brain regions in the A+ relative to the A-group only at M24. Theta power increase over the mid-frontal and mid-posterior cortices suggests an hypoactivation of the default-mode network in the A+ individuals and a non-linear longitudinal progression at M24. CONCLUSION We provide the first source-level longitudinal evidence on the impact of brain amyloidosis on the EEG dynamics of a large-scale, monocentric cohort of elderly individuals at-risk for AD.
Collapse
Affiliation(s)
- Giuseppe Spinelli
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Centre MEG-EEG, CENIR, Paris, France.,AP-HP, Hôpital de la Pitié-Salpêtrière, Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Paris, France
| | - Hovagim Bakardjian
- AP-HP, Hôpital de la Pitié-Salpêtrière, Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Paris, France
| | | | - Marie-Claude Potier
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Centre MEG-EEG, CENIR, Paris, France
| | - Marie-Odile Habert
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, Paris, France.,AP-HP, Hôpital de la Pitié-Salpêtrière, Médecine Nucléaire, Paris, France.,Centre d'Acquisition et Traitement des Images (CATI), http://www.cati-neuroimaging.com
| | - Marcel Levy
- AP-HP, Hôpital de la Pitié-Salpêtrière, Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Paris, France
| | - Bruno Dubois
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Centre MEG-EEG, CENIR, Paris, France.,AP-HP, Hôpital de la Pitié-Salpêtrière, Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Paris, France
| | - Nathalie George
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Centre MEG-EEG, CENIR, Paris, France
| | | |
Collapse
|
41
|
Xicota L, Gyorgy B, Grenier-Boley B, Lecoeur A, Fontaine G, Danjou F, Gonzalez JS, Colliot O, Amouyel P, Martin G, Levy M, Villain N, Habert MO, Dubois B, Lambert JC, Potier MC. Association of APOE-Independent Alzheimer Disease Polygenic Risk Score With Brain Amyloid Deposition in Asymptomatic Older Adults. Neurology 2022; 99:e462-e475. [PMID: 35606148 PMCID: PMC9421597 DOI: 10.1212/wnl.0000000000200544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 03/02/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Brain amyloid deposition, a major risk factor for Alzheimer disease (AD), is currently estimated by measuring CSF or plasma amyloid peptide levels or by PET imaging. Assessing genetic risks relating to amyloid deposition before any accumulation has occurred would allow for earlier intervention in persons at increased risk for developing AD. Previous work linking amyloid burden and genetic risk relied almost exclusively on APOE, a major AD genetic risk factor. Here, we ask whether a polygenic risk score (PRS) that incorporates an optimized list of common variants linked to AD and excludes APOE is associated with brain amyloid load in cognitively unimpaired older adults. METHODS We included 291 asymptomatic older participants from the INveStIGation of AlzHeimer's PredicTors (INSIGHT pre-AD) cohort who underwent amyloid imaging, including 83 amyloid-positive (+) participants. We used an Alzheimer's (A) PRS composed of 33 AD risk variants excluding APOE and selected the 17 variants that showed the strongest association with amyloid positivity to define an optimized (oA) PRS. Participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI) study (228 participants, 90 amyloid [+]) were tested as a validation cohort. Finally, 2,300 patients with AD and 6,994 controls from the European Alzheimer's Disease Initiative (EADI) were evaluated. RESULTS A-PRS was not significantly associated with amyloid burden in the INSIGHT or ADNI cohorts with or without correction for the APOE genotype. However, oA-PRS was significantly associated with amyloid status independently of APOE adjustment (INSIGHT odds ratio [OR]: 5.26 [1.71-16.88]; ADNI OR: 3.38 [1.02-11.63]). Of interest, oA-PRS accurately discriminated amyloid (+) and (-) APOE ε4 carriers (INSIGHT OR: 181.6 [7.53-10,674.6]; ADNI OR: 44.94 [3.03-1,277]). A-PRS and oA-PRS showed a significant association with disease status in the EADI cohort (OR: 1.68 [1.53-1.85] and 2.06 [1.73-2.45], respectively). Genes assigned to oA-PRS variants were enriched in ontologies related to β-amyloid metabolism and deposition. DISCUSSION PRSs relying on AD genetic risk factors excluding APOE may improve risk prediction for brain amyloid, allowing stratification of cognitively unimpaired individuals at risk of AD independent of their APOE status.
Collapse
Affiliation(s)
- Laura Xicota
- From the ICM Paris Brain Institute (L.X., B.G., A.L., G.«F., F.D., J.S.G., O.C., N.V., B.D., M.-C.P.), CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière; Univ. Lille (B.G.-B., P.A., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement; Inria (J.S.G., O.C.), Aramis-Project Team, Paris; Centre d'Acquisition et Traitement des Images (CATI platform) (G.M., M.-O.H.), cati-neuroimaging.com, Paris; Centre des Maladies Cognitives et Comportementales (M.L., M.-O.H., B.D.), IM2A, AP-HP, Sorbonne Université, Hôpital de la Salpêtrière; Department of Neurology (N.V., B.D.), Hôpital Pitié-Salpêtrière, AP-HP Sorbonne Université; Sorbonne Université (M.-O.H.), CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB; and AP-HP (M.-O.H.), Hôpital Pitié-Salpêtrière, Médecine Nucléaire, Paris, France
| | - Beata Gyorgy
- From the ICM Paris Brain Institute (L.X., B.G., A.L., G.«F., F.D., J.S.G., O.C., N.V., B.D., M.-C.P.), CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière; Univ. Lille (B.G.-B., P.A., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement; Inria (J.S.G., O.C.), Aramis-Project Team, Paris; Centre d'Acquisition et Traitement des Images (CATI platform) (G.M., M.-O.H.), cati-neuroimaging.com, Paris; Centre des Maladies Cognitives et Comportementales (M.L., M.-O.H., B.D.), IM2A, AP-HP, Sorbonne Université, Hôpital de la Salpêtrière; Department of Neurology (N.V., B.D.), Hôpital Pitié-Salpêtrière, AP-HP Sorbonne Université; Sorbonne Université (M.-O.H.), CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB; and AP-HP (M.-O.H.), Hôpital Pitié-Salpêtrière, Médecine Nucléaire, Paris, France
| | - Benjamin Grenier-Boley
- From the ICM Paris Brain Institute (L.X., B.G., A.L., G.«F., F.D., J.S.G., O.C., N.V., B.D., M.-C.P.), CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière; Univ. Lille (B.G.-B., P.A., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement; Inria (J.S.G., O.C.), Aramis-Project Team, Paris; Centre d'Acquisition et Traitement des Images (CATI platform) (G.M., M.-O.H.), cati-neuroimaging.com, Paris; Centre des Maladies Cognitives et Comportementales (M.L., M.-O.H., B.D.), IM2A, AP-HP, Sorbonne Université, Hôpital de la Salpêtrière; Department of Neurology (N.V., B.D.), Hôpital Pitié-Salpêtrière, AP-HP Sorbonne Université; Sorbonne Université (M.-O.H.), CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB; and AP-HP (M.-O.H.), Hôpital Pitié-Salpêtrière, Médecine Nucléaire, Paris, France
| | - Alexandre Lecoeur
- From the ICM Paris Brain Institute (L.X., B.G., A.L., G.«F., F.D., J.S.G., O.C., N.V., B.D., M.-C.P.), CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière; Univ. Lille (B.G.-B., P.A., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement; Inria (J.S.G., O.C.), Aramis-Project Team, Paris; Centre d'Acquisition et Traitement des Images (CATI platform) (G.M., M.-O.H.), cati-neuroimaging.com, Paris; Centre des Maladies Cognitives et Comportementales (M.L., M.-O.H., B.D.), IM2A, AP-HP, Sorbonne Université, Hôpital de la Salpêtrière; Department of Neurology (N.V., B.D.), Hôpital Pitié-Salpêtrière, AP-HP Sorbonne Université; Sorbonne Université (M.-O.H.), CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB; and AP-HP (M.-O.H.), Hôpital Pitié-Salpêtrière, Médecine Nucléaire, Paris, France
| | - Gaëlle Fontaine
- From the ICM Paris Brain Institute (L.X., B.G., A.L., G.«F., F.D., J.S.G., O.C., N.V., B.D., M.-C.P.), CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière; Univ. Lille (B.G.-B., P.A., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement; Inria (J.S.G., O.C.), Aramis-Project Team, Paris; Centre d'Acquisition et Traitement des Images (CATI platform) (G.M., M.-O.H.), cati-neuroimaging.com, Paris; Centre des Maladies Cognitives et Comportementales (M.L., M.-O.H., B.D.), IM2A, AP-HP, Sorbonne Université, Hôpital de la Salpêtrière; Department of Neurology (N.V., B.D.), Hôpital Pitié-Salpêtrière, AP-HP Sorbonne Université; Sorbonne Université (M.-O.H.), CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB; and AP-HP (M.-O.H.), Hôpital Pitié-Salpêtrière, Médecine Nucléaire, Paris, France
| | - Fabrice Danjou
- From the ICM Paris Brain Institute (L.X., B.G., A.L., G.«F., F.D., J.S.G., O.C., N.V., B.D., M.-C.P.), CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière; Univ. Lille (B.G.-B., P.A., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement; Inria (J.S.G., O.C.), Aramis-Project Team, Paris; Centre d'Acquisition et Traitement des Images (CATI platform) (G.M., M.-O.H.), cati-neuroimaging.com, Paris; Centre des Maladies Cognitives et Comportementales (M.L., M.-O.H., B.D.), IM2A, AP-HP, Sorbonne Université, Hôpital de la Salpêtrière; Department of Neurology (N.V., B.D.), Hôpital Pitié-Salpêtrière, AP-HP Sorbonne Université; Sorbonne Université (M.-O.H.), CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB; and AP-HP (M.-O.H.), Hôpital Pitié-Salpêtrière, Médecine Nucléaire, Paris, France
| | - Jorge Samper Gonzalez
- From the ICM Paris Brain Institute (L.X., B.G., A.L., G.«F., F.D., J.S.G., O.C., N.V., B.D., M.-C.P.), CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière; Univ. Lille (B.G.-B., P.A., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement; Inria (J.S.G., O.C.), Aramis-Project Team, Paris; Centre d'Acquisition et Traitement des Images (CATI platform) (G.M., M.-O.H.), cati-neuroimaging.com, Paris; Centre des Maladies Cognitives et Comportementales (M.L., M.-O.H., B.D.), IM2A, AP-HP, Sorbonne Université, Hôpital de la Salpêtrière; Department of Neurology (N.V., B.D.), Hôpital Pitié-Salpêtrière, AP-HP Sorbonne Université; Sorbonne Université (M.-O.H.), CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB; and AP-HP (M.-O.H.), Hôpital Pitié-Salpêtrière, Médecine Nucléaire, Paris, France
| | - Olivier Colliot
- From the ICM Paris Brain Institute (L.X., B.G., A.L., G.«F., F.D., J.S.G., O.C., N.V., B.D., M.-C.P.), CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière; Univ. Lille (B.G.-B., P.A., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement; Inria (J.S.G., O.C.), Aramis-Project Team, Paris; Centre d'Acquisition et Traitement des Images (CATI platform) (G.M., M.-O.H.), cati-neuroimaging.com, Paris; Centre des Maladies Cognitives et Comportementales (M.L., M.-O.H., B.D.), IM2A, AP-HP, Sorbonne Université, Hôpital de la Salpêtrière; Department of Neurology (N.V., B.D.), Hôpital Pitié-Salpêtrière, AP-HP Sorbonne Université; Sorbonne Université (M.-O.H.), CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB; and AP-HP (M.-O.H.), Hôpital Pitié-Salpêtrière, Médecine Nucléaire, Paris, France
| | - Philippe Amouyel
- From the ICM Paris Brain Institute (L.X., B.G., A.L., G.«F., F.D., J.S.G., O.C., N.V., B.D., M.-C.P.), CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière; Univ. Lille (B.G.-B., P.A., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement; Inria (J.S.G., O.C.), Aramis-Project Team, Paris; Centre d'Acquisition et Traitement des Images (CATI platform) (G.M., M.-O.H.), cati-neuroimaging.com, Paris; Centre des Maladies Cognitives et Comportementales (M.L., M.-O.H., B.D.), IM2A, AP-HP, Sorbonne Université, Hôpital de la Salpêtrière; Department of Neurology (N.V., B.D.), Hôpital Pitié-Salpêtrière, AP-HP Sorbonne Université; Sorbonne Université (M.-O.H.), CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB; and AP-HP (M.-O.H.), Hôpital Pitié-Salpêtrière, Médecine Nucléaire, Paris, France
| | - Garance Martin
- From the ICM Paris Brain Institute (L.X., B.G., A.L., G.«F., F.D., J.S.G., O.C., N.V., B.D., M.-C.P.), CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière; Univ. Lille (B.G.-B., P.A., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement; Inria (J.S.G., O.C.), Aramis-Project Team, Paris; Centre d'Acquisition et Traitement des Images (CATI platform) (G.M., M.-O.H.), cati-neuroimaging.com, Paris; Centre des Maladies Cognitives et Comportementales (M.L., M.-O.H., B.D.), IM2A, AP-HP, Sorbonne Université, Hôpital de la Salpêtrière; Department of Neurology (N.V., B.D.), Hôpital Pitié-Salpêtrière, AP-HP Sorbonne Université; Sorbonne Université (M.-O.H.), CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB; and AP-HP (M.-O.H.), Hôpital Pitié-Salpêtrière, Médecine Nucléaire, Paris, France
| | - Marcel Levy
- From the ICM Paris Brain Institute (L.X., B.G., A.L., G.«F., F.D., J.S.G., O.C., N.V., B.D., M.-C.P.), CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière; Univ. Lille (B.G.-B., P.A., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement; Inria (J.S.G., O.C.), Aramis-Project Team, Paris; Centre d'Acquisition et Traitement des Images (CATI platform) (G.M., M.-O.H.), cati-neuroimaging.com, Paris; Centre des Maladies Cognitives et Comportementales (M.L., M.-O.H., B.D.), IM2A, AP-HP, Sorbonne Université, Hôpital de la Salpêtrière; Department of Neurology (N.V., B.D.), Hôpital Pitié-Salpêtrière, AP-HP Sorbonne Université; Sorbonne Université (M.-O.H.), CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB; and AP-HP (M.-O.H.), Hôpital Pitié-Salpêtrière, Médecine Nucléaire, Paris, France
| | - Nicolas Villain
- From the ICM Paris Brain Institute (L.X., B.G., A.L., G.«F., F.D., J.S.G., O.C., N.V., B.D., M.-C.P.), CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière; Univ. Lille (B.G.-B., P.A., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement; Inria (J.S.G., O.C.), Aramis-Project Team, Paris; Centre d'Acquisition et Traitement des Images (CATI platform) (G.M., M.-O.H.), cati-neuroimaging.com, Paris; Centre des Maladies Cognitives et Comportementales (M.L., M.-O.H., B.D.), IM2A, AP-HP, Sorbonne Université, Hôpital de la Salpêtrière; Department of Neurology (N.V., B.D.), Hôpital Pitié-Salpêtrière, AP-HP Sorbonne Université; Sorbonne Université (M.-O.H.), CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB; and AP-HP (M.-O.H.), Hôpital Pitié-Salpêtrière, Médecine Nucléaire, Paris, France
| | - Marie-Odile Habert
- From the ICM Paris Brain Institute (L.X., B.G., A.L., G.«F., F.D., J.S.G., O.C., N.V., B.D., M.-C.P.), CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière; Univ. Lille (B.G.-B., P.A., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement; Inria (J.S.G., O.C.), Aramis-Project Team, Paris; Centre d'Acquisition et Traitement des Images (CATI platform) (G.M., M.-O.H.), cati-neuroimaging.com, Paris; Centre des Maladies Cognitives et Comportementales (M.L., M.-O.H., B.D.), IM2A, AP-HP, Sorbonne Université, Hôpital de la Salpêtrière; Department of Neurology (N.V., B.D.), Hôpital Pitié-Salpêtrière, AP-HP Sorbonne Université; Sorbonne Université (M.-O.H.), CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB; and AP-HP (M.-O.H.), Hôpital Pitié-Salpêtrière, Médecine Nucléaire, Paris, France
| | - Bruno Dubois
- From the ICM Paris Brain Institute (L.X., B.G., A.L., G.«F., F.D., J.S.G., O.C., N.V., B.D., M.-C.P.), CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière; Univ. Lille (B.G.-B., P.A., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement; Inria (J.S.G., O.C.), Aramis-Project Team, Paris; Centre d'Acquisition et Traitement des Images (CATI platform) (G.M., M.-O.H.), cati-neuroimaging.com, Paris; Centre des Maladies Cognitives et Comportementales (M.L., M.-O.H., B.D.), IM2A, AP-HP, Sorbonne Université, Hôpital de la Salpêtrière; Department of Neurology (N.V., B.D.), Hôpital Pitié-Salpêtrière, AP-HP Sorbonne Université; Sorbonne Université (M.-O.H.), CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB; and AP-HP (M.-O.H.), Hôpital Pitié-Salpêtrière, Médecine Nucléaire, Paris, France
| | - Jean-Charles Lambert
- From the ICM Paris Brain Institute (L.X., B.G., A.L., G.«F., F.D., J.S.G., O.C., N.V., B.D., M.-C.P.), CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière; Univ. Lille (B.G.-B., P.A., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement; Inria (J.S.G., O.C.), Aramis-Project Team, Paris; Centre d'Acquisition et Traitement des Images (CATI platform) (G.M., M.-O.H.), cati-neuroimaging.com, Paris; Centre des Maladies Cognitives et Comportementales (M.L., M.-O.H., B.D.), IM2A, AP-HP, Sorbonne Université, Hôpital de la Salpêtrière; Department of Neurology (N.V., B.D.), Hôpital Pitié-Salpêtrière, AP-HP Sorbonne Université; Sorbonne Université (M.-O.H.), CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB; and AP-HP (M.-O.H.), Hôpital Pitié-Salpêtrière, Médecine Nucléaire, Paris, France
| | - Marie-Claude Potier
- From the ICM Paris Brain Institute (L.X., B.G., A.L., G.«F., F.D., J.S.G., O.C., N.V., B.D., M.-C.P.), CNRS UMR7225, INSERM U1127, Sorbonne University, Hôpital de la Pitié-Salpêtrière; Univ. Lille (B.G.-B., P.A., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement; Inria (J.S.G., O.C.), Aramis-Project Team, Paris; Centre d'Acquisition et Traitement des Images (CATI platform) (G.M., M.-O.H.), cati-neuroimaging.com, Paris; Centre des Maladies Cognitives et Comportementales (M.L., M.-O.H., B.D.), IM2A, AP-HP, Sorbonne Université, Hôpital de la Salpêtrière; Department of Neurology (N.V., B.D.), Hôpital Pitié-Salpêtrière, AP-HP Sorbonne Université; Sorbonne Université (M.-O.H.), CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB; and AP-HP (M.-O.H.), Hôpital Pitié-Salpêtrière, Médecine Nucléaire, Paris, France.
| |
Collapse
|
42
|
Jiang Y, Wang Z, Liu X, Wan M, Liu Y, Jiao B, Liao X, Luo Z, Wang Y, Hong C, Tan Y, Weng L, Zhou Y, Rao S, Cao J, Liu Z, Wan T, Zhu Y, Xie H, Shen L. The Protective Effects of Osteocyte-Derived Extracellular Vesicles Against Alzheimer's Disease Diminished with Aging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105316. [PMID: 35508803 PMCID: PMC9189667 DOI: 10.1002/advs.202105316] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/16/2022] [Indexed: 05/16/2023]
Abstract
Both Alzheimer's disease (AD) and osteoporosis (OP) are common age-associated degenerative diseases and are strongly correlated with clinical epidemiology. However, there is a lack of clear pathological relationship between the brain and bone in the current understanding. Here, it is found that young osteocyte, the most abundant cells in bone, secretes extracellular vesicles (OCYYoung -EVs) to ameliorate cognitive impairment and the pathogenesis of AD in APP/PS1 mice and model cells. These benefits of OCYYoung -EVs are diminished in aged osteocyte-derived EVs (OCYAged -EVs). Based on the self-constructed OCY-EVs tracer transgenic mouse models and the in vivo fluorescent imaging system, OCY-EVs have been observed to be transported to the brain under physiological and pathological conditions. In the hippocampal administration of Aβ40 induced young AD model mice, the intramedullary injection of Rab27a-shRNA adenovirus inhibits OCYYoung -EVs secretion from bone and aggravates cognitive impairment. Proteomic quantitative analysis reveals that OCYYoung -EVs, compared to OCYAged -EVs, enrich multiple protective factors of AD pathway. The study uncovers the role of OCY-EV as a regulator of brain health, suggesting a novel mechanism in bone-brain communication.
Collapse
|
43
|
Grober E, Lipton RB, Sperling RA, Papp KV, Johnson KA, Rentz DM, Veroff AE, Aisen PS, Ezzati A. Associations of Stages of Objective Memory Impairment With Amyloid PET and Structural MRI: The A4 Study. Neurology 2022; 98:e1327-e1336. [PMID: 35197359 PMCID: PMC8967421 DOI: 10.1212/wnl.0000000000200046] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 01/03/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES The goal of this work was to investigate the neuroimaging correlates of the Stages of Objective Memory Impairment (SOMI) system operationalized with the Free and Cued Selective Reminding Test (FCSRT), a widely used episodic memory measure. METHODS The FCSRT begins with a study phase in which items (e.g., grapes) are identified in response to unique semantic cues (e.g., fruit) that are used in the test phase to prompt recall of items not retrieved by free recall. There are 3 test trials of the 16 items (maximum 48). Data from 4,484 cognitively unimpaired participants from the Anti-Amyloid Treatment in Asymptomatic Alzheimer's (A4) study were used. All participants had amyloid PET imaging, and a subset of 1,262 β-amyloid (Aβ)-positive had structural MRIs. We compared the Aβ mean cortical standardized uptake value ratio (SUVR) and volumetric measures of hippocampus, parahippocampal gyrus, entorhinal cortex, and inferior temporal cortex between the 5 SOMI stages. RESULTS Participants had a mean age of 71.3 (SD 4.6) years; 40.6% were male; and 34.6% were APOE ε4 positive. Half had no memory impairment; the other half had retrieval deficits, storage limitations, or both. Analysis of covariance in the entire sample while controlling for age, sex, education, and APOE ε4 showed that individuals in higher SOMI stages had higher global amyloid SUVR (p < 0.001). Both SOMI-4 and -3 subgroups had higher amyloid SUVR than SOMI-0 and SOMI-1 subgroups. Individuals in higher SOMI stages had smaller hippocampal volume (p = 0.003), entorhinal cortex (p < 0.05), and inferior temporal lobes (p < 0.05), but there was no difference between parahippocampal gyrus volume of different SOMI stages. Pairwise comparison of SOMI subgroups showed that the SOMI-4, -3, and -2 subgroups had smaller hippocampal volume than the SOMI-0 and -1 subgroup. The SOMI-4 subgroup had significantly smaller entorhinal cortex and smaller inferior temporal lobe compared to all other groups. DISCUSSION Presence of Alzheimer disease pathology is closely related to memory impairment according to SOMI stages in the cognitively unimpaired sample of A4. Results from structural MRIs suggest that memory storage impairment (SOMI-3 and -4) is present when there is widespread medial temporal lobe atrophy. TRIAL REGISTRATION INFORMATION ClinicalTrials.gov identifier: NCT02008357. CLASSIFICATION OF EVIDENCE This study provides Class I evidence that, in normal older individuals, higher stages of memory impairment assessed with FCSRT were associated with higher amyloid imaging burden and lower volume of hippocampus, entorhinal cortex, and inferior temporal lobes.
Collapse
Affiliation(s)
- Ellen Grober
- From the Department of Neurology (E.G., R.B.L., A.E.V., A.E.), Albert Einstein College of Medicine, Bronx, NY; Harvard Aging Brain Study (R.A.S., K.V.P., K.A.J., D.M.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School; Center for Alzheimer Research and Treatment (R.A.S., K.V.P., K.A.J., D.M.R.), Department of Neurology, Brigham and Women's Hospital, Boston, MA; and Alzheimer's Therapeutic Research Institute (P.S.A.), University of Southern California, San Diego.
| | - Richard B Lipton
- From the Department of Neurology (E.G., R.B.L., A.E.V., A.E.), Albert Einstein College of Medicine, Bronx, NY; Harvard Aging Brain Study (R.A.S., K.V.P., K.A.J., D.M.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School; Center for Alzheimer Research and Treatment (R.A.S., K.V.P., K.A.J., D.M.R.), Department of Neurology, Brigham and Women's Hospital, Boston, MA; and Alzheimer's Therapeutic Research Institute (P.S.A.), University of Southern California, San Diego
| | - Reisa A Sperling
- From the Department of Neurology (E.G., R.B.L., A.E.V., A.E.), Albert Einstein College of Medicine, Bronx, NY; Harvard Aging Brain Study (R.A.S., K.V.P., K.A.J., D.M.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School; Center for Alzheimer Research and Treatment (R.A.S., K.V.P., K.A.J., D.M.R.), Department of Neurology, Brigham and Women's Hospital, Boston, MA; and Alzheimer's Therapeutic Research Institute (P.S.A.), University of Southern California, San Diego
| | - Kathryn V Papp
- From the Department of Neurology (E.G., R.B.L., A.E.V., A.E.), Albert Einstein College of Medicine, Bronx, NY; Harvard Aging Brain Study (R.A.S., K.V.P., K.A.J., D.M.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School; Center for Alzheimer Research and Treatment (R.A.S., K.V.P., K.A.J., D.M.R.), Department of Neurology, Brigham and Women's Hospital, Boston, MA; and Alzheimer's Therapeutic Research Institute (P.S.A.), University of Southern California, San Diego
| | - Keith A Johnson
- From the Department of Neurology (E.G., R.B.L., A.E.V., A.E.), Albert Einstein College of Medicine, Bronx, NY; Harvard Aging Brain Study (R.A.S., K.V.P., K.A.J., D.M.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School; Center for Alzheimer Research and Treatment (R.A.S., K.V.P., K.A.J., D.M.R.), Department of Neurology, Brigham and Women's Hospital, Boston, MA; and Alzheimer's Therapeutic Research Institute (P.S.A.), University of Southern California, San Diego
| | - Dorene M Rentz
- From the Department of Neurology (E.G., R.B.L., A.E.V., A.E.), Albert Einstein College of Medicine, Bronx, NY; Harvard Aging Brain Study (R.A.S., K.V.P., K.A.J., D.M.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School; Center for Alzheimer Research and Treatment (R.A.S., K.V.P., K.A.J., D.M.R.), Department of Neurology, Brigham and Women's Hospital, Boston, MA; and Alzheimer's Therapeutic Research Institute (P.S.A.), University of Southern California, San Diego
| | - Amy E Veroff
- From the Department of Neurology (E.G., R.B.L., A.E.V., A.E.), Albert Einstein College of Medicine, Bronx, NY; Harvard Aging Brain Study (R.A.S., K.V.P., K.A.J., D.M.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School; Center for Alzheimer Research and Treatment (R.A.S., K.V.P., K.A.J., D.M.R.), Department of Neurology, Brigham and Women's Hospital, Boston, MA; and Alzheimer's Therapeutic Research Institute (P.S.A.), University of Southern California, San Diego
| | - Paul S Aisen
- From the Department of Neurology (E.G., R.B.L., A.E.V., A.E.), Albert Einstein College of Medicine, Bronx, NY; Harvard Aging Brain Study (R.A.S., K.V.P., K.A.J., D.M.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School; Center for Alzheimer Research and Treatment (R.A.S., K.V.P., K.A.J., D.M.R.), Department of Neurology, Brigham and Women's Hospital, Boston, MA; and Alzheimer's Therapeutic Research Institute (P.S.A.), University of Southern California, San Diego
| | - Ali Ezzati
- From the Department of Neurology (E.G., R.B.L., A.E.V., A.E.), Albert Einstein College of Medicine, Bronx, NY; Harvard Aging Brain Study (R.A.S., K.V.P., K.A.J., D.M.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School; Center for Alzheimer Research and Treatment (R.A.S., K.V.P., K.A.J., D.M.R.), Department of Neurology, Brigham and Women's Hospital, Boston, MA; and Alzheimer's Therapeutic Research Institute (P.S.A.), University of Southern California, San Diego
| |
Collapse
|
44
|
Li TR, Yang Q, Hu X, Han Y. Biomarkers and Tools for Predicting Alzheimer's Disease in the Preclinical Stage. Curr Neuropharmacol 2022; 20:713-737. [PMID: 34030620 PMCID: PMC9878962 DOI: 10.2174/1570159x19666210524153901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 04/27/2021] [Accepted: 05/08/2021] [Indexed: 11/22/2022] Open
Abstract
Alzheimer's disease (AD) is the only leading cause of death for which no disease-modifying therapy is currently available. Over the past decade, a string of disappointing clinical trial results has forced us to shift our focus to the preclinical stage of AD, which represents the most promising therapeutic window. However, the accurate diagnosis of preclinical AD requires the presence of brain β- amyloid deposition determined by cerebrospinal fluid or amyloid-positron emission tomography, significantly limiting routine screening and diagnosis in non-tertiary hospital settings. Thus, an easily accessible marker or tool with high sensitivity and specificity is highly needed. Recently, it has been discovered that individuals in the late stage of preclinical AD may not be truly "asymptomatic" in that they may have already developed subtle or subjective cognitive decline. In addition, advances in bloodderived biomarker studies have also allowed the detection of pathologic changes in preclinical AD. Exosomes, as cell-to-cell communication messengers, can reflect the functional changes of their source cell. Methodological advances have made it possible to extract brain-derived exosomes from peripheral blood, making exosomes an emerging biomarker carrier and liquid biopsy tool for preclinical AD. The eye and its associated structures have rich sensory-motor innervation. In this regard, studies have indicated that they may also provide reliable markers. Here, our report covers the current state of knowledge of neuropsychological and eye tests as screening tools for preclinical AD and assesses the value of blood and brain-derived exosomes as carriers of biomarkers in conjunction with the current diagnostic paradigm.
Collapse
Affiliation(s)
- Tao-Ran Li
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Qin Yang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Xiaochen Hu
- Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, 50924, Germany
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China;,Center of Alzheimer’s Disease, Beijing Institute for Brain Disorders, Beijing, 100053, China;,National Clinical Research Center for Geriatric Disorders, Beijing, 100053, China;,School of Biomedical Engineering, Hainan University, Haikou, 570228, China;,Address correspondence to this author at the Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China; Tel: +86 13621011941; E-mail:
| |
Collapse
|
45
|
Kok FK, van Leerdam SL, de Lange ECM. Potential Mechanisms Underlying Resistance to Dementia in Non-Demented Individuals with Alzheimer's Disease Neuropathology. J Alzheimers Dis 2022; 87:51-81. [PMID: 35275527 PMCID: PMC9198800 DOI: 10.3233/jad-210607] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Alzheimer’s disease (AD) is the most common form of dementia and typically characterized by the accumulation of amyloid-β plaques and tau tangles. Intriguingly, there also exists a group of elderly which do not develop dementia during their life, despite the AD neuropathology, the so-called non-demented individuals with AD neuropathology (NDAN). In this review, we provide extensive background on AD pathology and normal aging and discuss potential mechanisms that enable these NDAN individuals to remain cognitively intact. Studies presented in this review show that NDAN subjects are generally higher educated and have a larger cognitive reserve. Furthermore, enhanced neural hypertrophy could compensate for hippocampal and cingulate neural atrophy in NDAN individuals. On a cellular level, these individuals show increased levels of neural stem cells and ‘von Economo neurons’. Furthermore, in NDAN brains, binding of Aβ oligomers to synapses is prevented, resulting in decreased glial activation and reduced neuroinflammation. Overall, the evidence stated here strengthens the idea that some individuals are more resistant to AD pathology, or at least show an elongation of the asymptomatic state of the disease compared to others. Insights into the mechanisms underlying this resistance could provide new insight in understanding normal aging and AD itself. Further research should focus on factors and mechanisms that govern the NDAN cognitive resilience in order to find clues on novel biomarkers, targets, and better treatments of AD.
Collapse
Affiliation(s)
- Frédérique K Kok
- Predictive Pharmacology, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Suzanne L van Leerdam
- Predictive Pharmacology, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Elizabeth C M de Lange
- Predictive Pharmacology, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
46
|
Zhang SS, Zhu L, Peng Y, Zhang L, Chao FL, Jiang L, Xiao Q, Liang X, Tang J, Yang H, He Q, Guo YJ, Zhou CN, Tang Y. Long-term running exercise improves cognitive function and promotes microglial glucose metabolism and morphological plasticity in the hippocampus of APP/PS1 mice. J Neuroinflammation 2022; 19:34. [PMID: 35123512 PMCID: PMC8817568 DOI: 10.1186/s12974-022-02401-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 01/24/2022] [Indexed: 02/06/2023] Open
Abstract
Background The role of physical exercise in the prevention of Alzheimer’s disease (AD) has been widely studied. Microglia play an important role in AD. Triggering receptor expressed in myeloid cells 2 (TREM2) is expressed on microglia and is known to mediate microglial metabolic activity and brain glucose metabolism. However, the relationship between brain glucose metabolism and microglial metabolic activity during running exercise in APP/PS1 mice remains unclear. Methods Ten-month-old male APP/PS1 mice and wild-type mice were randomly divided into sedentary groups or running groups (AD_Sed, WT_Sed, AD_Run and WT_Run, n = 20/group). Running mice had free access to a running wheel for 3 months. Behavioral tests, [18]F-FDG-PET and hippocampal RNA-Seq were performed. The expression levels of microglial glucose transporter (GLUT5), TREM2, soluble TREM2 (sTREM2), TYRO protein tyrosine kinase binding protein (TYROBP), secreted phosphoprotein 1 (SPP1), and phosphorylated spleen tyrosine kinase (p-SYK) were estimated by western blot or ELISA. Immunohistochemistry, stereological methods and immunofluorescence were used to investigate the morphology, proliferation and activity of microglia. Results Long-term voluntary running significantly improved cognitive function in APP/PS1 mice. Although there were few differentially expressed genes (DEGs), gene set enrichment analysis (GSEA) showed enriched glycometabolic pathways in APP/PS1 running mice. Running exercise increased FDG uptake in the hippocampus of APP/PS1 mice, as well as the protein expression of GLUT5, TREM2, SPP1 and p-SYK. The level of sTREM2 decreased in the plasma of APP/PS1 running mice. The number of microglia, the length and endpoints of microglial processes, and the ratio of GLUT5+/IBA1+ microglia were increased in the dentate gyrus (DG) of APP/PS1 running mice. Running exercise did not alter the number of 5-bromo-2′-deoxyuridine (BrdU)+/IBA1+ microglia but reduced the immunoactivity of CD68 in the hippocampus of APP/PS1 mice. Conclusions Running exercise inhibited TREM2 shedding and maintained TREM2 protein levels, which were accompanied by the promotion of brain glucose metabolism, microglial glucose metabolism and morphological plasticity in the hippocampus of AD mice. Microglia might be a structural target responsible for the benefits of running exercise in AD. Promoting microglial glucose metabolism and morphological plasticity modulated by TREM2 might be a novel strategy for AD treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02401-5.
Collapse
|
47
|
Jack CR, Therneau TM, Lundt ES, Wiste HJ, Mielke MM, Knopman DS, Graff-Radford J, Lowe VJ, Vemuri P, Schwarz CG, Senjem ML, Gunter JL, Petersen RC. Long-term associations between amyloid positron emission tomography, sex, apolipoprotein E and incident dementia and mortality among individuals without dementia: hazard ratios and absolute risk. Brain Commun 2022; 4:fcac017. [PMID: 35310829 PMCID: PMC8924651 DOI: 10.1093/braincomms/fcac017] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/08/2021] [Accepted: 01/31/2022] [Indexed: 11/14/2022] Open
Abstract
Dementia and mortality rates rise inexorably with age and consequently interact. However, because of the major logistical difficulties in accounting for both outcomes in a defined population, very little work has examined how risk factors and biomarkers for incident dementia are influenced by competing mortality. The objective of this study was to examine long-term associations between amyloid PET, APOE ɛ4, sex, education and cardiovascular/metabolic conditions, and hazard and absolute risk of dementia and mortality in individuals without dementia at enrolment. Participants were enrolled in the Mayo Clinic Study of Aging, a population-based study of cognitive ageing in Olmsted County, MN, USA. All were without dementia and were age 55-92 years at enrolment and were followed longitudinally. Predictor variables were amyloid PET, APOE ɛ4 status, sex, education, cardiovascular/metabolic conditions and age. The main outcomes were incident dementia and mortality. Multivariable, multi-state models were used to estimate mortality and incident dementia rates and absolute risk of dementia and mortality by predictor variable group. Of the 4984 participants in the study, 4336 (87%) were cognitively unimpaired and 648 (13%) had mild cognitive impairment at enrolment. The median age at enrolment was 75 years; 2463 (49%) were women. The median follow-up time was 9.4 years (7.5 years after PET). High versus normal amyloid (hazard ratio 2.11, 95% confidence interval 1.43-2.79), APOE ɛ4 (women: hazard ratio 2.24, 95% confidence interval 1.80-2.77; men: hazard ratio 1.37, 95% confidence interval 1.09-1.71), older age and two additional cardiovascular/metabolic conditions (hazard ratio 1.37, 95% confidence interval 1.22-1.53) were associated with the increased hazard of dementia (all P < 0.001). Among APOE ɛ4 carriers with elevated amyloid, remaining lifetime risk of dementia at age 65 years was greater in women [74% (95% confidence interval 65-84%) high and 58% (95% confidence interval 52-65%) moderate amyloid], than men [62% (95% confidence interval 52-73%) high and 44% (95% confidence interval 35-53%) moderate amyloid]. Overall, the hazard and absolute risk of dementia varied considerably by predictor group. The absolute risk of dementia associated with predictors characteristic of Alzheimer's disease was greater in women than men while at the same time the combination of APOE ɛ4 non-carrier with normal amyloid was more protective in women than men. This set of findings may be attributed in part to different biological effects and in part to lower mortality rates in women.
Collapse
Affiliation(s)
| | - Terry M. Therneau
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Emily S. Lundt
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Heather J. Wiste
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Michelle M. Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Val J. Lowe
- Department of Nuclear Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | |
Collapse
|
48
|
Mahaman YAR, Embaye KS, Huang F, Li L, Zhu F, Wang JZ, Liu R, Feng J, Wang X. Biomarkers used in Alzheimer's disease diagnosis, treatment, and prevention. Ageing Res Rev 2022; 74:101544. [PMID: 34933129 DOI: 10.1016/j.arr.2021.101544] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 12/09/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD), being the number one in terms of dementia burden, is an insidious age-related neurodegenerative disease and is presently considered a global public health threat. Its main histological hallmarks are the Aβ senile plaques and the P-tau neurofibrillary tangles, while clinically it is marked by a progressive cognitive decline that reflects the underlying synaptic loss and neurodegeneration. Many of the drug therapies targeting the two pathological hallmarks namely Aβ and P-tau have been proven futile. This is probably attributed to the initiation of therapy at a stage where cognitive alterations are already obvious. In other words, the underlying neuropathological changes are at a stage where these drugs lack any therapeutic value in reversing the damage. Therefore, there is an urgent need to start treatment in the very early stage where these changes can be reversed, and hence, early diagnosis is of primordial importance. To this aim, the use of robust and informative biomarkers that could provide accurate diagnosis preferably at an earlier phase of the disease is of the essence. To date, several biomarkers have been established that, to a different extent, allow researchers and clinicians to evaluate, diagnose, and more specially exclude other related pathologies. In this study, we extensively reviewed data on the currently explored biomarkers in terms of AD pathology-specific and non-specific biomarkers and highlighted the recent developments in the diagnostic and theragnostic domains. In the end, we have presented a separate elaboration on aspects of future perspectives and concluding remarks.
Collapse
|
49
|
Gagliardi G, Vannini P. Episodic Memory Impairment Mediates the Loss of Awareness in Mild Cognitive Impairment. Front Aging Neurosci 2022; 13:802501. [PMID: 35126092 PMCID: PMC8814670 DOI: 10.3389/fnagi.2021.802501] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/30/2021] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION Loss of awareness is a common symptom in Alzheimer's Disease (AD) and responsible for a significant loss of functional abilities. The mechanisms underlying loss of awareness in AD is unknown, although previous findings have implicated dysfunction of primary executive functioning (EF) or episodic memory (EM) to be the cause. Therefore, our main study objective was to explore the involvement of EF and EM dysfunction in amyloid-related loss of awareness across the clinical spectrum of AD. METHODS A total of 895 participants (362 clinically normal [CN], 422 people with mild cognitive impairment [MCI] and 111 with dementia) from the Alzheimer's Disease Neuroimaging Initiative were used for the analyses. A sub-analysis was performed in 202 participants who progressed in their clinical diagnosis from CN to MCI or MCI to dementia as well as dementia patients. Mediation models were used in each clinical group with awareness (assessed with the Everyday Cognitive function questionnaire) as a dependent variable to determine whether EF and/or EM would mediate the effect of amyloid on awareness. We also ran these analyses with subjective and informant complaints as dependent variables. Direct correlations between all variables were also performed. RESULTS We found evidence for a decline in awareness across the groups, with increased awareness observed in the CN group and decreased awareness observed in the MCI and dementia groups. Our results showed that EM, and not EF, partially mediated the relationship between amyloid and awareness such that greater amyloid and lower EM performance was associated with lower awareness. When analyzing each group separately, this finding was only observed in the MCI group and in the group containing progressors and dementia patients. When repeating the analyses for subjective and informant complaints separately, the results were replicated only for the informant's complaints. DISCUSSION Our results demonstrate that decline in EM and, to a lesser degree, EF, mediate the effect of amyloid on awareness. In line with previous studies demonstrating the development of anosognosia in the prodromal stage, our findings suggest that decreased awareness is the result of an inability for the participant to update his/her insight into his/her cognitive performance (i.e., demonstrating a petrified self).
Collapse
Affiliation(s)
- Geoffroy Gagliardi
- Neurology, Brigham and Women's Hospital, Boston, MA, United States
- Neurology, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Cambridge, MA, United States
| | - Patrizia Vannini
- Neurology, Brigham and Women's Hospital, Boston, MA, United States
- Neurology, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Cambridge, MA, United States
| |
Collapse
|
50
|
de Mendonça Lima CA. Towards a WPA Position Document on the Human Rights of Older Adults with Mental Health Conditions: К документу с изложением позиции Всемирной психиатрической ассоциации по вопросу соблюдения прав пожилых людей с психическими расстройствами. CONSORTIUM PSYCHIATRICUM 2022; 3:16-21. [PMID: 39045355 PMCID: PMC11262087 DOI: 10.17816/cp150] [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: 02/01/2022] [Accepted: 02/12/2022] [Indexed: 11/08/2022] Open
Abstract
The increasing number of older adults in countries across the world is a huge challenge to those that are in charge of promoting, protecting, and implementing their human rights. This task is particularly difficult in the absence of a strong international framework addressing the principles required to guide the actions to combat all human rights violations. The existence of such a specific framework for older adults with mental health conditions is justified in view of the particular vulnerability of this section of the population by virtue of societal ageism, stigmatization, exclusion, as well as the disability and dependency which mental health conditions in old age may confer. The present article is a development of a previous statement by the International Psychogeriatric Association and the World Psychiatric Association Section of Old Age Psychiatry. As there is a call to all organizations to support efforts to combat Human Rights violations among older adults, a text will be submitted to the Executive Committee of the World Psychiatric Association to approve an official position statement on Human Rights of Older Persons with Mental Health Conditions.
Collapse
|