1
|
van den Berg RL, Butterbrod E, de Boer C, Schlüter L, van Harten AC, Teunissen CE, van de Giessen E, van der Flier WM, Sikkes SAM. Amyloid-related changes in fluency in patients with subjective cognitive decline. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2025; 17:e70063. [PMID: 39822289 PMCID: PMC11736636 DOI: 10.1002/dad2.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 10/25/2024] [Accepted: 12/06/2024] [Indexed: 01/19/2025]
Abstract
INTRODUCTION We examined semantic and phonemic fluency in individuals with subjective cognitive decline (SCD) in relation to amyloid status and clinical progression. METHODS A total of 490 individuals with SCD (62 ± 8 years, 42% female, 28% amyloid-positive, 17% clinical progression) completed annual fluency assessments (mean ± SD follow-up 4.3 ± 2.9 years). Associations between fluency trajectories, amyloid status, and clinical progression were examined with linear mixed models and joint models. RESULTS Amyloid-positive individuals declined faster than amyloid-negative individuals on semantic fluency (B = -0.35, p < 0.001), but not on phonemic fluency (B = -0.06, p = 0.218). An annual decline of one word in semantic and phonemic fluency was associated with 22% (hazard ratio [HR] = 1.22, p < 0.001) and 28% (HR = 1.28, p = 0.004) increased risk of clinical progression. DISCUSSION Our results indicate that decline in semantic fluency is an early indicator of cognitive deficits in preclinical Alzheimer's disease. Highlights Abnormal amyloid burden is associated with decline in semantic fluency.Fluency trajectories are associated with an increased risk of clinical progression.More refined measures are needed to detect the earliest language deficits.
Collapse
Affiliation(s)
- Rosanne L. van den Berg
- Alzheimer Center Amsterdam, NeurologyAmsterdam University Medical CenterAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
- Department of Clinical, Neuro and Developmental Psychology, Faculty of Movement and Behavioral SciencesVU UniversityAmsterdamthe Netherlands
| | - Elke Butterbrod
- Alzheimer Center Amsterdam, NeurologyAmsterdam University Medical CenterAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
- Department of Clinical, Neuro and Developmental Psychology, Faculty of Movement and Behavioral SciencesVU UniversityAmsterdamthe Netherlands
- Department of NeurosurgeryElisabeth‐Tweesteden HospitalTilburgthe Netherlands
| | - Casper de Boer
- Alzheimer Center Amsterdam, NeurologyAmsterdam University Medical CenterAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Lisa‐Marie Schlüter
- Alzheimer Center Amsterdam, NeurologyAmsterdam University Medical CenterAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Argonde C. van Harten
- Alzheimer Center Amsterdam, NeurologyAmsterdam University Medical CenterAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Charlotte E. Teunissen
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
- Department of Clinical ChemistryNeurochemistry Laboratory and BiobankAmsterdam NeuroscienceAmsterdam University Medical CenterAmsterdamthe Netherlands
| | - Elsmarieke van de Giessen
- Department of Radiology & Nuclear MedicineAmsterdam University Medical CenterAmsterdamthe Netherlands
- Amsterdam Neuroscience, Brain ImagingAmsterdamthe Netherlands
| | - Wiesje M. van der Flier
- Alzheimer Center Amsterdam, NeurologyAmsterdam University Medical CenterAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
- Department of Epidemiology and BiostatisticsAmsterdam NeuroscienceVU University Medical CenterAmsterdamthe Netherlands
| | - Sietske A. M. Sikkes
- Alzheimer Center Amsterdam, NeurologyAmsterdam University Medical CenterAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
- Department of Clinical, Neuro and Developmental Psychology, Faculty of Movement and Behavioral SciencesVU UniversityAmsterdamthe Netherlands
| |
Collapse
|
2
|
Ataka T, Kimura N, Kaneko N, Masuda T, Takeuchi Y, Yabuuchi K, Mizukami T, Takeuchi T, Ito T, Tasai H, Miyagawa T, Hanai S, Iwamoto S, Matsubara E. Plasma amyloid beta biomarkers predict amyloid positivity and longitudinal clinical progression in mild cognitive impairment. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e70008. [PMID: 39748849 PMCID: PMC11694533 DOI: 10.1002/trc2.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 01/04/2025]
Abstract
INTRODUCTION Previous studies have examined the predictive accuracy of plasma amyloid beta (Aβ) biomarkers in clinical cohorts. However, their accuracy for predicting amyloid-positive patients in community-based cohorts is unclear. This study aimed to determine the predictive accuracy of Aβ precursor protein 669-711/Aβ1-42, Aβ1-40/1-42 and their composite biomarkers for brain amyloid deposition or the clinical progression in community-dwelling older adults with mild cognitive impairment (MCI). METHODS This prospective cohort study was conducted from August 2015 to September 2019. Subsequently, the participants underwent follow-up cognitive assessments up to 8 years after the start of the study. Blood samples were collected from older adults aged ≥ 65 years with MCI at baseline. Plasma Aβ biomarkers were analyzed using immunoprecipitation-mass spectrometry. The accuracy of plasma biomarkers for brain amyloid status was evaluated using receiver operating characteristic curve analysis. Relationships between comorbidities and plasma Aβ markers were examined using multiple linear regression analysis. Associations of plasma biomarkers with clinical conversion to Alzheimer's disease (AD) dementia were evaluated using Kaplan‒Meier curves. RESULTS The participants included 107 patients (57 [53.3%] females, median age: 76.0 [72.0-80.0] years). Plasma biomarkers correlated with cortical amyloid uptake (ρ = 0.667-0.754). The composite biomarker had the best area under the curve (0.943, 95% confidence interval [CI]: 0.901 to 0.985) for predicting amyloid positivity. Apolipoprotein ε4 status showed significant correlations with increased plasma amyloid biomarker levels. Participants with high composite biomarker levels at baseline had a greater risk of conversion to AD dementia (hazard ratio 10.74, 95% CI: 3.51 to 32.84, P < 0.001). The higher composite biomarker was associated with a faster rate of cognitive decline (ρ = -0.575, P < 0.001). DISCUSSION Plasma Aβ composite biomarker may serve as a surrogate measure for amyloid deposition and a predictor of disease progression in a community-based cohort. Highlights Plasma amyloid beta (Aβ) biomarkers correlated with 11C-Pittsburgh compound B uptake, mainly in the frontal/parietotemporal cortices and posterior cingulate gyrus.The amyloid composite biomarker can predict amyloid positron emission tomography positivity with a high area under the curve of 0.943 in a community-based mild cognitive impairment cohort.The higher amyloid composite biomarker at baseline was significantly associated with worsening Mini-Mental State Examination score and a high risk for developing Alzheimer's disease (AD) dementia over 8 years.The amyloid composite biomarker can predict clinical progression to AD dementia with a high area under the curve of 0.860.Apolipoprotein E ε4 status influenced the plasma Aβ biomarker levels.
Collapse
Affiliation(s)
- Takuya Ataka
- Department of NeurologyFaculty of MedicineOita UniversityOitaJapan
| | - Noriyuki Kimura
- Department of NeurologyFaculty of MedicineOita UniversityOitaJapan
| | - Naoki Kaneko
- Koichi Tanaka Mass Spectrometry Research LaboratoryShimadzu CorporationKyotoJapan
| | - Teruaki Masuda
- Department of NeurologyFaculty of MedicineOita UniversityOitaJapan
| | - Yosuke Takeuchi
- Department of NeurologyFaculty of MedicineOita UniversityOitaJapan
| | - Kenichi Yabuuchi
- Department of NeurologyFaculty of MedicineOita UniversityOitaJapan
| | - Takeshi Mizukami
- Department of NeurologyFaculty of MedicineOita UniversityOitaJapan
| | - Tsukasa Takeuchi
- Koichi Tanaka Mass Spectrometry Research LaboratoryShimadzu CorporationKyotoJapan
| | | | | | | | - Shunya Hanai
- Koichi Tanaka Mass Spectrometry Research LaboratoryShimadzu CorporationKyotoJapan
| | - Shinichi Iwamoto
- Koichi Tanaka Mass Spectrometry Research LaboratoryShimadzu CorporationKyotoJapan
| | - Etsuro Matsubara
- Department of NeurologyFaculty of MedicineOita UniversityOitaJapan
| |
Collapse
|
3
|
Shao K, Hu X, Kleineidam L, Stark M, Altenstein S, Amthauer H, Boecker H, Buchert R, Buerger K, Butryn M, Cai Y, Cai Y, Cosma NC, Chen G, Chen Z, Daamen M, Drzezga A, Düzel E, Essler M, Ewers M, Fliessbach K, Gaertner FC, Glanz W, Guo T, Hansen N, He B, Janowitz D, Kilimann I, Krause BJ, Lan G, Lange C, Laske C, Li Y, Li R, Liu L, Lu J, Meng F, Munk MH, Peters O, Perneczky R, Priller J, Ramirez A, Rauchmann B, Reimold M, Rominger A, Rostamzadeh A, Roy‐Kluth N, Schneider A, Spottke A, Spruth EJ, Sun P, Teipel S, Wang X, Wei M, Wei Y, Wiltfang J, Yan S, Yang J, Yu X, Zhang M, Zhang L, Wagner M, Jessen F, Han Y, Kuhn E. Amyloid and SCD jointly predict cognitive decline across Chinese and German cohorts. Alzheimers Dement 2024; 20:5926-5939. [PMID: 39072956 PMCID: PMC11497667 DOI: 10.1002/alz.14119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/10/2024] [Accepted: 05/11/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION Subjective cognitive decline (SCD) in amyloid-positive (Aβ+) individuals was proposed as a clinical indicator of Stage 2 in the Alzheimer's disease (AD) continuum, but this requires further validation across cultures, measures, and recruitment strategies. METHODS Eight hundred twenty-one participants from SILCODE and DELCODE cohorts, including normal controls (NC) and individuals with SCD recruited from the community or from memory clinics, underwent neuropsychological assessments over up to 6 years. Amyloid positivity was derived from positron emission tomography or plasma biomarkers. Global cognitive change was analyzed using linear mixed-effects models. RESULTS In the combined and stratified cohorts, Aβ+ participants with SCD showed steeper cognitive decline or diminished practice effects compared with NC or Aβ- participants with SCD. These findings were confirmed using different operationalizations of SCD and amyloid positivity, and across different SCD recruitment settings. DISCUSSION Aβ+ individuals with SCD in German and Chinese populations showed greater global cognitive decline and could be targeted for interventional trials. HIGHLIGHTS SCD in amyloid-positive (Aβ+) participants predicts a steeper cognitive decline. This finding does not rely on specific SCD or amyloid operationalization. This finding is not specific to SCD patients recruited from memory clinics. This finding is valid in both German and Chinese populations. Aβ+ older adults with SCD could be a target population for interventional trials.
Collapse
Affiliation(s)
- Kai Shao
- Department of NeurologyXuanWu Hospital of Capital Medical UniversityBeijingChina
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of PsychiatryMedical FacultyUniversity of CologneCologneGermany
| | - Xiaochen Hu
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of PsychiatryMedical FacultyUniversity of CologneCologneGermany
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Old Age Psychiatry and Cognitive DisordersUniversity of Bonn Medical CenterBonnGermany
| | - Melina Stark
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Old Age Psychiatry and Cognitive DisordersUniversity of Bonn Medical CenterBonnGermany
| | - Slawek Altenstein
- German Center for Neurodegenerative Diseases (DZNE)BerlinGermany
- Department of Psychiatry and PsychotherapyCharitéBerlinGermany
| | - Holger Amthauer
- Department of Nuclear MedicineCharité ‐ Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, and Humboldt‐Universität zu BerlinBerlinGermany
| | - Henning Boecker
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Clinical Functional Imaging Group, Department of Diagnostic and Interventional RadiologyUniversity Hospital BonnBonnGermany
| | - Ralph Buchert
- Department of Nuclear MedicineCharité ‐ Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, and Humboldt‐Universität zu Berlin and Berlin Institute of HealthBerlinGermany
- Department of Diagnostic and Interventional Radiology and Nuclear MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Katharina Buerger
- German Center for Neurodegenerative Diseases (DZNE, Munich)MunichGermany
- Institute for Stroke and Dementia Research (ISD)University Hospital, LMU MunichMunichGermany
| | - Michaela Butryn
- German Center for Neurodegenerative Diseases (DZNE)MagdeburgGermany
- Institute of Cognitive Neurology and Dementia Research (IKND)Otto‐von‐Guericke UniversityMagdeburgGermany
| | - Yanning Cai
- Department of clinical biobankXuanWu Hospital of Capital Medical UniversityBeijingChina
| | - Yue Cai
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
| | - Nicoleta Carmen Cosma
- Department of Psychiatry and PsychotherapyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Guanqun Chen
- Department of NeurologyBeijing ChaoYang Hospital of Capital Medical UniversityBeijingChina
| | - Zhigeng Chen
- Department of Radiology and Nuclear MedicineXuanWu Hospital of Capital Medical UniversityBeijingChina
| | - Marcel Daamen
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Alexander Drzezga
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
- Institute of Neuroscience and Medicine (INM‐2)Molecular Organization of the Brain, Forschungszentrum JülichJülichGermany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE)MagdeburgGermany
- Institute of Cognitive Neurology and Dementia Research (IKND)Otto‐von‐Guericke UniversityMagdeburgGermany
| | - Markus Essler
- Department of Nuclear MedicineUniversity Hospital BonnBonnGermany
| | - Michael Ewers
- German Center for Neurodegenerative Diseases (DZNE, Munich)MunichGermany
- Institute for Stroke and Dementia Research (ISD)University Hospital, LMU MunichMunichGermany
| | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Old Age Psychiatry and Cognitive DisordersUniversity of Bonn Medical CenterBonnGermany
| | | | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE)MagdeburgGermany
- Institute of Cognitive Neurology and Dementia Research (IKND)Otto‐von‐Guericke UniversityMagdeburgGermany
| | - Tengfei Guo
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
| | - Niels Hansen
- Department of Psychiatry and Psychotherapy, University Medical Center GoettingenUniversity of GoettingenGoettingenGermany
| | - Beiqi He
- School of Information and Communication EngineeringHainan UniversityHaikouChina
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD)University Hospital, LMU MunichMunichGermany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE)RostockGermany
- Department of Psychosomatic MedicineRostock University Medical CenterRostockGermany
| | - Bernd J. Krause
- Department of Nuclear MedicineRostock University Medical CentreRostockGermany
| | - Guoyu Lan
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
- Tsinghua Shenzhen International Graduate School (SIGS)Tsinghua UniversityShenzhenChina
| | - Catharina Lange
- Department of Nuclear MedicineCharité ‐ Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, and Humboldt‐Universität zu BerlinBerlinGermany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and PsychotherapyUniversity of TübingenTübingenGermany
| | - Yuxia Li
- Department of NeurologyTangshan Central HospitalTanshanChina
| | - Ruixian Li
- Department of NeurologyXuanWu Hospital of Capital Medical UniversityBeijingChina
| | - Lin Liu
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
- Tsinghua Shenzhen International Graduate School (SIGS)Tsinghua UniversityShenzhenChina
| | - Jie Lu
- Department of Radiology and Nuclear MedicineXuanWu Hospital of Capital Medical UniversityBeijingChina
| | - Fansheng Meng
- Medical Imaging Department of Hainan Cancer HospitalHaikouChina
| | - Matthias H. Munk
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
- Department of Psychiatry and PsychotherapyUniversity of TübingenTübingenGermany
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE)BerlinGermany
- Department of Psychiatry and PsychotherapyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE, Munich)MunichGermany
- Department of Psychiatry and PsychotherapyUniversity Hospital, LMU MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy) MunichMunichGermany
- Ageing Epidemiology Research Unit (AGE), School of Public HealthImperial College LondonLondonUK
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE)BerlinGermany
- Department of Psychiatry and PsychotherapyCharitéBerlinGermany
- University of Edinburgh and UK DRIEdinburghUK
- School of Medicine, Department of Psychiatry and PsychotherapyTechnical University of MunichMunichGermany
| | - Alfredo Ramirez
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Old Age Psychiatry and Cognitive DisordersUniversity of Bonn Medical CenterBonnGermany
- Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneKölnGermany
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital CologneUniversity of CologneKölnGermany
- Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesSan AntonioTexasUSA
| | - Boris‐Stephan Rauchmann
- Department of Psychiatry and PsychotherapyUniversity Hospital, LMU MunichMunichGermany
- Sheffield Institute for Translational Neuroscience (SITraN)University of SheffieldSheffieldUK
- Department of NeuroradiologyUniversity Hospital LMUMunichGermany
| | - Matthias Reimold
- Department of Nuclear Medicine and Clinical Molecular ImagingEberhard‐Karls‐UniversityTuebingenGermany
| | - Axel Rominger
- Department of Nuclear MedicineLudwig‐Maximilian‐University MunichMunichGermany
- Department of Nuclear Medicine, Inselspital, Bern University HospitalUniversity of BernBernSwitzerland
| | - Ayda Rostamzadeh
- Department of PsychiatryMedical FacultyUniversity of CologneCologneGermany
| | - Nina Roy‐Kluth
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Old Age Psychiatry and Cognitive DisordersUniversity of Bonn Medical CenterBonnGermany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of NeurologyUniversity of BonnBonnGermany
| | - Eike Jakob Spruth
- German Center for Neurodegenerative Diseases (DZNE)BerlinGermany
- Department of Psychiatry and PsychotherapyCharitéBerlinGermany
| | - Pan Sun
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
- Tsinghua Shenzhen International Graduate School (SIGS)Tsinghua UniversityShenzhenChina
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE)RostockGermany
- Department of Psychosomatic MedicineRostock University Medical CenterRostockGermany
| | - Xiao Wang
- Department of Psychiatry and PsychotherapyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Min Wei
- Department of NeurologyXuanWu Hospital of Capital Medical UniversityBeijingChina
| | - Yongzhe Wei
- Department of NeurologyXuanWu Hospital of Capital Medical UniversityBeijingChina
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center GoettingenUniversity of GoettingenGoettingenGermany
- German Center for Neurodegenerative Diseases (DZNE)GoettingenGermany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical SciencesUniversity of AveiroAveiroPortugal
| | - Shaozhen Yan
- Department of Radiology and Nuclear MedicineXuanWu Hospital of Capital Medical UniversityBeijingChina
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
| | - Jie Yang
- Department of NeurologyXuanWu Hospital of Capital Medical UniversityBeijingChina
| | - Xianfeng Yu
- Department of NeurologyXuanWu Hospital of Capital Medical UniversityBeijingChina
| | - Mingkai Zhang
- Department of NeurologyXuanWu Hospital of Capital Medical UniversityBeijingChina
| | - Liang Zhang
- School of Information and Communication EngineeringHainan UniversityHaikouChina
| | | | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Old Age Psychiatry and Cognitive DisordersUniversity of Bonn Medical CenterBonnGermany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of PsychiatryMedical FacultyUniversity of CologneCologneGermany
- Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneKölnGermany
| | - Ying Han
- Department of NeurologyXuanWu Hospital of Capital Medical UniversityBeijingChina
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
- School of Biomedical EngineeringHainan UniversityHaikouChina
- Center of Alzheimer's DiseaseBeijing Institute for Brain DisordersBeijingChina
- National Clinical Research Center for Geriatric DisordersBeijingChina
- The Central Hospital of KaramayXinjiangChina
| | - Elizabeth Kuhn
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Old Age Psychiatry and Cognitive DisordersUniversity of Bonn Medical CenterBonnGermany
| |
Collapse
|
4
|
Zhang Y, Ferreira PCL, Jacobsen E, Bellaver B, Pascoal TA, Snitz BE, Chang CH, Villemagne VL, Ganguli M, Karikari TK. Association of plasma biomarkers of Alzheimer's disease and related disorders with cognition and cognitive decline: The MYHAT population-based study. Alzheimers Dement 2024; 20:4199-4211. [PMID: 38753951 PMCID: PMC11180930 DOI: 10.1002/alz.13829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 05/18/2024]
Abstract
INTRODUCTION Plasma biomarkers of Alzheimer's disease and related dementias predict global cognitive performance and decline over time; it remains unclear how they associate with changes in different dementia syndromes affecting distinct cognitive domains. METHODS In a prospective study with repeated assessments of a randomly selected population-based cohort (n = 787, median age 73), we evaluated performance and decline in different cognitive domains over up to 8 years in relation to plasma concentrations of amyloid beta 42/40 (Aβ42/40) ratio, phosphorylated tau181 (p-tau181), neurofilament light chain (NfL), and glial fibrillary acidic protein (GFAP). RESULTS Cross-sectionally, memory showed the strongest associations with p-tau181, and attention, executive, and visuospatial functions with NfL. Longitudinally, memory decline was distinguishable with all biomarker profiles dichotomized according to data-driven cutoffs, most efficiently with Aβ42/40. GFAP and Aβ42/40 were the best discriminators of decline patterns in language and visuospatial functions, respectively. DISCUSSION These relatively non-invasive tests may be beneficial for clinical screening after replication in other populations and validation through neuroimaging or cerebrospinal fluid analysis. HIGHLIGHTS We performed a prospective study with up to 8 years of repeated domain-specific cognitive assessments and baseline plasma Alzheimer's disease and related dementias biomarker measurements in a randomly selected population-based cohort. We considered distinct growth curves of trajectories of different cognitive domains and survival bias induced by missing data by adding quadratic time and applying joint modeling technique. Cross-sectionally, memory showed the strongest associations with plasma phosphorylated tau181, while attention, executive, and visuospatial functions were most strongly associated with neurofilament light chain. Longitudinally, memory and visuospatial declines were most efficiently distinguished by dichotomized amyloid beta 42/40 profile among all plasma biomarkers, while language was by dichotomized glial fibrillary acidic protein. These relatively non-invasive tests may be beneficial for clinical screening; however, they will need replication in other populations and validation through neuroimaging and/or cerebrospinal fluid assessments.
Collapse
Affiliation(s)
- Yingjin Zhang
- Department of Biostatistics School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Pamela C. L. Ferreira
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Erin Jacobsen
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Bruna Bellaver
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Tharick A. Pascoal
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of NeurologySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Beth E. Snitz
- Department of NeurologySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Chung‐Chou H. Chang
- Department of Biostatistics School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of MedicineSchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Victor L. Villemagne
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Mary Ganguli
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of NeurologySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Epidemiology School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Thomas K. Karikari
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
5
|
Grasset L, Bouteloup V, Cacciamani F, Pellegrin I, Planche V, Chêne G, Dufouil C. Associations Between Blood-Based Biomarkers and Cognitive and Functional Trajectories Among Participants of the MEMENTO Cohort. Neurology 2024; 102:e209307. [PMID: 38626384 PMCID: PMC11175638 DOI: 10.1212/wnl.0000000000209307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/05/2024] [Indexed: 04/18/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Elevated levels of Alzheimer disease (AD) blood-based biomarkers are associated with accelerated cognitive decline. However, their distinct relationships with specific cognitive and functional domains require further investigation. We aimed at estimating the associations between AD blood-based biomarkers and the trajectories of distinct cognitive and functional domains over a 5-year follow-up period. METHODS We conducted a clinic-based prospective study using data from the MEMENTO study, a nationwide French cohort. We selected dementia-free individuals at baseline aged 60 years or older. Baseline measurements of β-amyloid (Aβ) 40 and 42, phosphorylated tau (p-tau181), and neurofilament light chain (NfL) concentrations were obtained using the Simoa HD-X analyzer. Mini-Mental State Examination (MMSE), Free and Cued Selective Reminding Test (FCSRT), animal fluency, Trail Making Tests A and B, Short Physical Performance Battery (SPPB), and Instrumental Activities of Daily Living were administered annually for up to 5 years. We used linear mixed models, adjusted for potential confounders, to model AD biomarkers' relation with cognitive and functional decline. RESULTS A total of 1,938 participants were included in this study, with a mean (SD) baseline age of 72.8 (6.6) years, and 62% were women. Higher baseline p-tau181 and NfL were associated with significantly faster decline in most cognitive, physical, and functional outcomes (+1 SD p-tau181: βMMSE = -0.055, 95% CI -0.067 to -0.043, βFCSRT = -0.034, 95% CI -0.043 to -0.025, βfluency = -0.029, 95% CI -0.038 to -0.020, βSPPB = -0.040, 95% CI -0.057 to -0.022, and β4IADL = -0.115, 95% CI 0.091-0.140. +1 SD NfL: βMMSE = -0.039, 95% CI -0.053 to -0.025, βFCSRT = -0.022, 95% CI -0.032 to -0.012, βfluency = -0.014, 95% CI -0.024 to -0.004, and β4IADL = 0.077, 95% CI 0.048-0.105). A multiplicative association of p-tau181 and NfL with worsening cognitive and functional trajectories was evidenced. Lower Aβ42/40 ratio was only associated with slightly faster cognitive decline in FCSRT and semantic fluency (+1 SD: β = 0.011, 95% CI 0.002-0.020, and β = 0.011, 95% CI 0.003-0.020, respectively). These associations were not modified by APOE ε4, sex, nor education level. DISCUSSION In a memory clinic sample, p-tau181 and NfL, both independently and jointly, are linked to more pronounced cognitive, physical and functional declines. Blood-based biomarker measurement in AD research may provide useful insights regarding biological processes underlying cognitive, physical, and functional declines in at-risk individuals.
Collapse
Affiliation(s)
- Leslie Grasset
- From the UMR 1219 (L.G., V.B., F.C., G.C., C.D.), Bordeaux Population Health Center, University of Bordeaux, Inserm; CIC 1401-EC (L.G., V.B., F.C., G.C., C.D.), Inserm, University of Bordeaux, CHU de Bordeaux; Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), Pole de sante publique; ARAMISLab (F.C.), Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière; Qairnel SAS (F.C.), Paris; Laboratory of Immunology and Immunogenetics (I.P.), Resources Biological Center (CRB), CHU Bordeaux; Univ. Bordeaux (I.P.), CNRS, ImmunoConcEpT, UMR 5164; and Univ. Bordeaux (V.P.), CNRS UMR 5293, Institut des Maladies Neurodégénératives, Centre Mémoire de Ressources et de Recherches, Pôle de Neurosciences Cliniques, CHU de Bordeaux, France
| | - Vincent Bouteloup
- From the UMR 1219 (L.G., V.B., F.C., G.C., C.D.), Bordeaux Population Health Center, University of Bordeaux, Inserm; CIC 1401-EC (L.G., V.B., F.C., G.C., C.D.), Inserm, University of Bordeaux, CHU de Bordeaux; Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), Pole de sante publique; ARAMISLab (F.C.), Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière; Qairnel SAS (F.C.), Paris; Laboratory of Immunology and Immunogenetics (I.P.), Resources Biological Center (CRB), CHU Bordeaux; Univ. Bordeaux (I.P.), CNRS, ImmunoConcEpT, UMR 5164; and Univ. Bordeaux (V.P.), CNRS UMR 5293, Institut des Maladies Neurodégénératives, Centre Mémoire de Ressources et de Recherches, Pôle de Neurosciences Cliniques, CHU de Bordeaux, France
| | - Federica Cacciamani
- From the UMR 1219 (L.G., V.B., F.C., G.C., C.D.), Bordeaux Population Health Center, University of Bordeaux, Inserm; CIC 1401-EC (L.G., V.B., F.C., G.C., C.D.), Inserm, University of Bordeaux, CHU de Bordeaux; Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), Pole de sante publique; ARAMISLab (F.C.), Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière; Qairnel SAS (F.C.), Paris; Laboratory of Immunology and Immunogenetics (I.P.), Resources Biological Center (CRB), CHU Bordeaux; Univ. Bordeaux (I.P.), CNRS, ImmunoConcEpT, UMR 5164; and Univ. Bordeaux (V.P.), CNRS UMR 5293, Institut des Maladies Neurodégénératives, Centre Mémoire de Ressources et de Recherches, Pôle de Neurosciences Cliniques, CHU de Bordeaux, France
| | - Isabelle Pellegrin
- From the UMR 1219 (L.G., V.B., F.C., G.C., C.D.), Bordeaux Population Health Center, University of Bordeaux, Inserm; CIC 1401-EC (L.G., V.B., F.C., G.C., C.D.), Inserm, University of Bordeaux, CHU de Bordeaux; Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), Pole de sante publique; ARAMISLab (F.C.), Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière; Qairnel SAS (F.C.), Paris; Laboratory of Immunology and Immunogenetics (I.P.), Resources Biological Center (CRB), CHU Bordeaux; Univ. Bordeaux (I.P.), CNRS, ImmunoConcEpT, UMR 5164; and Univ. Bordeaux (V.P.), CNRS UMR 5293, Institut des Maladies Neurodégénératives, Centre Mémoire de Ressources et de Recherches, Pôle de Neurosciences Cliniques, CHU de Bordeaux, France
| | - Vincent Planche
- From the UMR 1219 (L.G., V.B., F.C., G.C., C.D.), Bordeaux Population Health Center, University of Bordeaux, Inserm; CIC 1401-EC (L.G., V.B., F.C., G.C., C.D.), Inserm, University of Bordeaux, CHU de Bordeaux; Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), Pole de sante publique; ARAMISLab (F.C.), Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière; Qairnel SAS (F.C.), Paris; Laboratory of Immunology and Immunogenetics (I.P.), Resources Biological Center (CRB), CHU Bordeaux; Univ. Bordeaux (I.P.), CNRS, ImmunoConcEpT, UMR 5164; and Univ. Bordeaux (V.P.), CNRS UMR 5293, Institut des Maladies Neurodégénératives, Centre Mémoire de Ressources et de Recherches, Pôle de Neurosciences Cliniques, CHU de Bordeaux, France
| | - Geneviève Chêne
- From the UMR 1219 (L.G., V.B., F.C., G.C., C.D.), Bordeaux Population Health Center, University of Bordeaux, Inserm; CIC 1401-EC (L.G., V.B., F.C., G.C., C.D.), Inserm, University of Bordeaux, CHU de Bordeaux; Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), Pole de sante publique; ARAMISLab (F.C.), Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière; Qairnel SAS (F.C.), Paris; Laboratory of Immunology and Immunogenetics (I.P.), Resources Biological Center (CRB), CHU Bordeaux; Univ. Bordeaux (I.P.), CNRS, ImmunoConcEpT, UMR 5164; and Univ. Bordeaux (V.P.), CNRS UMR 5293, Institut des Maladies Neurodégénératives, Centre Mémoire de Ressources et de Recherches, Pôle de Neurosciences Cliniques, CHU de Bordeaux, France
| | - Carole Dufouil
- From the UMR 1219 (L.G., V.B., F.C., G.C., C.D.), Bordeaux Population Health Center, University of Bordeaux, Inserm; CIC 1401-EC (L.G., V.B., F.C., G.C., C.D.), Inserm, University of Bordeaux, CHU de Bordeaux; Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), Pole de sante publique; ARAMISLab (F.C.), Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière; Qairnel SAS (F.C.), Paris; Laboratory of Immunology and Immunogenetics (I.P.), Resources Biological Center (CRB), CHU Bordeaux; Univ. Bordeaux (I.P.), CNRS, ImmunoConcEpT, UMR 5164; and Univ. Bordeaux (V.P.), CNRS UMR 5293, Institut des Maladies Neurodégénératives, Centre Mémoire de Ressources et de Recherches, Pôle de Neurosciences Cliniques, CHU de Bordeaux, France
| |
Collapse
|
6
|
Toniolo S, Zhao S, Scholcz A, Amein B, Ganse‐Dumrath A, Heslegrave AJ, Thompson S, Manohar S, Zetterberg H, Husain M. Relationship of plasma biomarkers to digital cognitive tests in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12590. [PMID: 38623387 PMCID: PMC11016819 DOI: 10.1002/dad2.12590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/17/2024]
Abstract
INTRODUCTION A major limitation in Alzheimer's disease (AD) research is the lack of the ability to measure cognitive performance at scale-robustly, remotely, and frequently. Currently, there are no established online digital platforms validated against plasma biomarkers of AD. METHODS We used a novel web-based platform that assessed different cognitive functions in AD patients (N = 46) and elderly controls (N = 53) who were also evaluated for plasma biomarkers (amyloid beta 42/40 ratio, phosphorylated tau ([p-tau]181, glial fibrillary acidic protein, neurofilament light chain). Their cognitive performance was compared to a second, larger group of elderly controls (N = 352). RESULTS Patients with AD were significantly impaired across all digital cognitive tests, with performance correlating with plasma biomarker levels, particularly p-tau181. The combination of p-tau181 and the single best-performing digital test achieved high accuracy in group classification. DISCUSSION These findings show how online testing can now be deployed in patients with AD to measure cognitive function effectively and related to blood biomarkers of the disease. Highlights This is the first study comparing online digital testing to plasma biomarkers.Alzheimer's disease patients and two independent cohorts of elderly controls were assessed.Cognitive performance correlated with plasma biomarkers, particularly phosphorylated tau (p-tau)181.Glial fibrillary acidic protein and neurofilament light chain, and less so the amyloid beta 42/40 ratio, were also associated with performance.The best cognitive metric performed at par to p-tau181 in group classification.
Collapse
Affiliation(s)
- Sofia Toniolo
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
- Cognitive Disorders ClinicJR HospitalOxfordUK
| | - Sijia Zhao
- Department of Experimental PsychologyUniversity of OxfordOxfordUK
| | - Anna Scholcz
- Department of Experimental PsychologyUniversity of OxfordOxfordUK
| | - Benazir Amein
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Akke Ganse‐Dumrath
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Amanda J. Heslegrave
- UK Dementia Research InstituteUCLLondonUK
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
| | | | - Sanjay Manohar
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
- Cognitive Disorders ClinicJR HospitalOxfordUK
- Department of Experimental PsychologyUniversity of OxfordOxfordUK
| | - Henrik Zetterberg
- UK Dementia Research InstituteUCLLondonUK
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
- Institute of Neuroscience and PhysiologyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Masud Husain
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
- Cognitive Disorders ClinicJR HospitalOxfordUK
- Department of Experimental PsychologyUniversity of OxfordOxfordUK
| |
Collapse
|
7
|
Huang L, Li Q, Lu Y, Pan F, Cui L, Wang Y, Miao Y, Chen T, Li Y, Wu J, Chen X, Jia J, Guo Q. Consensus on rapid screening for prodromal Alzheimer's disease in China. Gen Psychiatr 2024; 37:e101310. [PMID: 38313393 PMCID: PMC10836380 DOI: 10.1136/gpsych-2023-101310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 12/19/2023] [Indexed: 02/06/2024] Open
Abstract
Alzheimer's disease (AD) is a common cause of dementia, characterised by cerebral amyloid-β deposition, pathological tau and neurodegeneration. The prodromal stage of AD (pAD) refers to patients with mild cognitive impairment (MCI) and evidence of AD's pathology. At this stage, disease-modifying interventions should be used to prevent the progression to dementia. Given the inherent heterogeneity of MCI, more specific biomarkers are needed to elucidate the underlying AD's pathology. Although the uses of cerebrospinal fluid and positron emission tomography are widely accepted methods for detecting AD's pathology, their clinical applications are limited by their high costs and invasiveness, particularly in low-income areas in China. Therefore, to improve the early detection of Alzheimer's disease (AD) pathology through cost-effective screening methods, a panel of 45 neurologists, psychiatrists and gerontologists was invited to establish a formal consensus on the screening of pAD in China. The supportive evidence and grades of recommendations are based on a systematic literature review and focus group discussion. National meetings were held to allow participants to review, vote and provide their expert opinions to reach a consensus. A majority (two-thirds) decision was used for questions for which consensus could not be reached. Recommended screening methods are presented in this publication, including neuropsychological assessment, peripheral biomarkers and brain imaging. In addition, a general workflow for screening pAD in China is established, which will help clinicians identify individuals at high risk and determine therapeutic targets.
Collapse
Affiliation(s)
- Lin Huang
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qinjie Li
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Lu
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fengfeng Pan
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Cui
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Wang
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ya Miao
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianlu Chen
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yatian Li
- Shanghai BestCovered, Shanghai, China
| | | | - Xiaochun Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jianping Jia
- Department of Neurology, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Qihao Guo
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Bhalala OG, Watson R, Yassi N. Multi-Omic Blood Biomarkers as Dynamic Risk Predictors in Late-Onset Alzheimer's Disease. Int J Mol Sci 2024; 25:1231. [PMID: 38279230 PMCID: PMC10816901 DOI: 10.3390/ijms25021231] [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: 12/07/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Late-onset Alzheimer's disease is the leading cause of dementia worldwide, accounting for a growing burden of morbidity and mortality. Diagnosing Alzheimer's disease before symptoms are established is clinically challenging, but would provide therapeutic windows for disease-modifying interventions. Blood biomarkers, including genetics, proteins and metabolites, are emerging as powerful predictors of Alzheimer's disease at various timepoints within the disease course, including at the preclinical stage. In this review, we discuss recent advances in such blood biomarkers for determining disease risk. We highlight how leveraging polygenic risk scores, based on genome-wide association studies, can help stratify individuals along their risk profile. We summarize studies analyzing protein biomarkers, as well as report on recent proteomic- and metabolomic-based prediction models. Finally, we discuss how a combination of multi-omic blood biomarkers can potentially be used in memory clinics for diagnosis and to assess the dynamic risk an individual has for developing Alzheimer's disease dementia.
Collapse
Affiliation(s)
- Oneil G. Bhalala
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; (R.W.); (N.Y.)
- Department of Neurology, Melbourne Brain Centre at The Royal Melbourne Hospital, University of Melbourne, Parkville 3050, Australia
| | - Rosie Watson
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; (R.W.); (N.Y.)
- Department of Medicine, The Royal Melbourne Hospital, University of Melbourne, Parkville 3050, Australia
| | - Nawaf Yassi
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; (R.W.); (N.Y.)
- Department of Neurology, Melbourne Brain Centre at The Royal Melbourne Hospital, University of Melbourne, Parkville 3050, Australia
- Department of Medicine, The Royal Melbourne Hospital, University of Melbourne, Parkville 3050, Australia
| |
Collapse
|
9
|
Colmant L, Boyer E, Gerard T, Sleegers K, Lhommel R, Ivanoiu A, Lefèvre P, Kienlen-Campard P, Hanseeuw B. Definition of a Threshold for the Plasma Aβ42/Aβ40 Ratio Measured by Single-Molecule Array to Predict the Amyloid Status of Individuals without Dementia. Int J Mol Sci 2024; 25:1173. [PMID: 38256246 PMCID: PMC10816992 DOI: 10.3390/ijms25021173] [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: 12/15/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by amyloid beta (Aβ) plaques and hyperphosphorylated tau in the brain. Aβ plaques precede cognitive impairments and can be detected through amyloid-positron emission tomography (PET) or in cerebrospinal fluid (CSF). Assessing the plasma Aβ42/Aβ40 ratio seems promising for non-invasive and cost-effective detection of brain Aβ accumulation. This approach involves some challenges, including the accuracy of blood-based biomarker measurements and the establishment of clear, standardized thresholds to categorize the risk of developing brain amyloid pathology. Plasma Aβ42/Aβ40 ratio was measured in 277 volunteers without dementia, 70 AD patients and 18 non-AD patients using single-molecule array. Patients (n = 88) and some volunteers (n = 66) were subject to evaluation of amyloid status by CSF Aβ quantification or PET analysis. Thresholds of plasma Aβ42/Aβ40 ratio were determined based on a Gaussian mixture model, a decision tree, and the Youden's index. The 0.0472 threshold, the one with the highest sensitivity, was retained for general population without dementia screening, and the 0.0450 threshold was retained for research and clinical trials recruitment, aiming to minimize the need for CSF or PET analyses to identify amyloid-positive individuals. These findings offer a promising step towards a cost-effective method for identifying individuals at risk of developing AD.
Collapse
Affiliation(s)
- Lise Colmant
- Institute of Neuroscience, UCLouvain, 1200 Brussels, Belgium; (L.C.); (E.B.); (T.G.); (P.L.); (P.K.-C.)
- Neurology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium;
- Institute of Information and Communication Technologies, Electronics and Applied Mathematics, UCLouvain, 1348 Louvain-la-Neuve, Belgium
| | - Emilien Boyer
- Institute of Neuroscience, UCLouvain, 1200 Brussels, Belgium; (L.C.); (E.B.); (T.G.); (P.L.); (P.K.-C.)
- Neurology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium;
| | - Thomas Gerard
- Institute of Neuroscience, UCLouvain, 1200 Brussels, Belgium; (L.C.); (E.B.); (T.G.); (P.L.); (P.K.-C.)
- Neurology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium;
| | - Kristel Sleegers
- Complex Genetics of Alzheimer’s Disease Group, VIB-UAntwerp Center for Molecular Neurology, University of Antwerp, 2000 Antwerpen, Belgium;
| | - Renaud Lhommel
- Neurology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium;
| | - Adrian Ivanoiu
- Institute of Neuroscience, UCLouvain, 1200 Brussels, Belgium; (L.C.); (E.B.); (T.G.); (P.L.); (P.K.-C.)
- Neurology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium;
| | - Philippe Lefèvre
- Institute of Neuroscience, UCLouvain, 1200 Brussels, Belgium; (L.C.); (E.B.); (T.G.); (P.L.); (P.K.-C.)
- Institute of Information and Communication Technologies, Electronics and Applied Mathematics, UCLouvain, 1348 Louvain-la-Neuve, Belgium
| | - Pascal Kienlen-Campard
- Institute of Neuroscience, UCLouvain, 1200 Brussels, Belgium; (L.C.); (E.B.); (T.G.); (P.L.); (P.K.-C.)
| | - Bernard Hanseeuw
- Institute of Neuroscience, UCLouvain, 1200 Brussels, Belgium; (L.C.); (E.B.); (T.G.); (P.L.); (P.K.-C.)
- Neurology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium;
- WELBIO Department, WEL Research Institute, Avenue Pasteur, 6, 1300 Wavre, Belgium
| |
Collapse
|
10
|
Huber H, Ashton NJ, Schieren A, Montoliu-Gaya L, Molfetta GD, Brum WS, Lantero-Rodriguez J, Grötschel L, Stoffel-Wagner B, Coenen M, Weinhold L, Schmid M, Blennow K, Stehle P, Zetterberg H, Simon MC. Levels of Alzheimer's disease blood biomarkers are altered after food intake-A pilot intervention study in healthy adults. Alzheimers Dement 2023; 19:5531-5540. [PMID: 37243891 DOI: 10.1002/alz.13163] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/29/2023]
Abstract
INTRODUCTION Blood biomarkers accurately identify Alzheimer's disease (AD) pathophysiology and axonal injury. We investigated the influence of food intake on AD-related biomarkers in cognitively healthy, obese adults at high metabolic risk. METHODS One-hundred eleven participants underwent repeated blood sampling during 3 h after a standardized meal (postprandial group, PG). For comparison, blood was sampled from a fasting subgroup over 3 h (fasting group, FG). Plasma neurofilament light (NfL), glial fibrillary acidic protein (GFAP), amyloid-beta (Aβ) 42/40, phosphorylated tau (p-tau) 181 and 231, and total-tau were measured via single molecule array assays. RESULTS Significant differences were found for NfL, GFAP, Aβ42/40, p-tau181, and p-tau231 between FG and PG. The greatest change to baseline occurred for GFAP and p-tau181 (120 min postprandially, p < 0.0001). CONCLUSION Our data suggest that AD-related biomarkers are altered by food intake. Further studies are needed to verify whether blood biomarker sampling should be performed in the fasting state. HIGHLIGHTS Acute food intake alters plasma biomarkers of Alzheimer's disease in obese, otherwise healthy adults. We also found dynamic fluctuations in plasma biomarkers concentration in the fasting state suggesting physiological diurnal variations. Further investigations are highly needed to verify if biomarker measurements should be performed in the fasting state and at a standardized time of day to improve the diagnostic accuracy.
Collapse
Affiliation(s)
- Hanna Huber
- Nutritional Physiology, Institute of Nutrition and Food Science, University of Bonn, Bonn, Germany
- Nutrition and Microbiota, Institute of Nutrition and Food Science, University of Bonn, Bonn, Germany
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, Mölndal, Sweden
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Alina Schieren
- Nutrition and Microbiota, Institute of Nutrition and Food Science, University of Bonn, Bonn, Germany
| | - Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, Mölndal, Sweden
| | - Guglielmo Di Molfetta
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, Mölndal, Sweden
| | - Wagner S Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, Mölndal, Sweden
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, Mölndal, Sweden
| | - Lana Grötschel
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, Mölndal, Sweden
| | - Birgit Stoffel-Wagner
- Central Laboratory, Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Martin Coenen
- Clinical Study Core Unit, Study Center Bonn, Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Leonie Weinhold
- Institute of Medical Biometry, Informatics and Epidemiology, University of Bonn, Bonn, Germany
| | - Matthias Schmid
- Institute of Medical Biometry, Informatics and Epidemiology, University of Bonn, Bonn, Germany
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal Hospital, Mölndal, Sweden
| | - Peter Stehle
- Nutritional Physiology, Institute of Nutrition and Food Science, University of Bonn, Bonn, Germany
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Marie-Christine Simon
- Nutrition and Microbiota, Institute of Nutrition and Food Science, University of Bonn, Bonn, Germany
| |
Collapse
|
11
|
Pais MV, Forlenza OV, Diniz BS. Plasma Biomarkers of Alzheimer's Disease: A Review of Available Assays, Recent Developments, and Implications for Clinical Practice. J Alzheimers Dis Rep 2023; 7:355-380. [PMID: 37220625 PMCID: PMC10200198 DOI: 10.3233/adr-230029] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 04/03/2023] [Indexed: 05/25/2023] Open
Abstract
Recently, low-sensitive plasma assays have been replaced by new ultra-sensitive assays such as single molecule enzyme-linked immunosorbent assay (Simoa), the Mesoscale Discovery (MSD) platform, and immunoprecipitation-mass spectrometry (IP-MS) with higher accuracy in the determination of plasma biomarkers of Alzheimer's disease (AD). Despite the significant variability, many studies have established in-house cut-off values for the most promising available biomarkers. We first reviewed the most used laboratory methods and assays to measure plasma AD biomarkers. Next, we review studies focused on the diagnostic performance of these biomarkers to identify AD cases, predict cognitive decline in pre-clinical AD cases, and differentiate AD cases from other dementia. We summarized data from studies published until January 2023. A combination of plasma Aβ42/40 ratio, age, and APOE status showed the best accuracy in diagnosing brain amyloidosis with a liquid chromatography-mass spectrometry (LC-MS) assay. Plasma p-tau217 has shown the best accuracy in distinguishing Aβ-PET+ from Aβ-PET-even in cognitively unimpaired individuals. We also summarized the different cut-off values for each biomarker when available. Recently developed assays for plasma biomarkers have undeniable importance in AD research, with improved analytical and diagnostic performance. Some biomarkers have been extensively used in clinical trials and are now clinically available. Nonetheless, several challenges remain to their widespread use in clinical practice.
Collapse
Affiliation(s)
- Marcos V. Pais
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Faculdade de Medicina, Universidade de Sao Paulo (FMUSP), Sao Paulo, SP, Brazil
| | - Orestes V. Forlenza
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Faculdade de Medicina, Universidade de Sao Paulo (FMUSP), Sao Paulo, SP, Brazil
| | - Breno S. Diniz
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
12
|
Saunders TS, Pozzolo FE, Heslegrave A, King D, McGeachan RI, Spires-Jones MP, Harris SE, Ritchie C, Muniz-Terrera G, Deary IJ, Cox SR, Zetterberg H, Spires-Jones TL. Predictive blood biomarkers and brain changes associated with age-related cognitive decline. Brain Commun 2023; 5:fcad113. [PMID: 37180996 PMCID: PMC10167767 DOI: 10.1093/braincomms/fcad113] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 12/28/2022] [Accepted: 04/05/2023] [Indexed: 04/08/2023] Open
Abstract
Growing evidence supports the use of plasma levels of tau phosphorylated at threonine 181, amyloid-β, neurofilament light and glial fibrillary acidic protein as promising biomarkers for Alzheimer's disease. While these blood biomarkers are promising for distinguishing people with Alzheimer's disease from healthy controls, their predictive validity for age-related cognitive decline without dementia remains unclear. Further, while tau phosphorylated at threonine 181 is a promising biomarker, the distribution of this phospho-epitope of tau in the brain is unknown. Here, we tested whether plasma levels of tau phosphorylated at threonine 181, amyloid-β, neurofilament light and fibrillary acidic protein predict cognitive decline between ages 72 and 82 in 195 participants in the Lothian birth cohorts 1936 study of cognitive ageing. We further examined post-mortem brain samples from temporal cortex to determine the distribution of tau phosphorylated at threonine 181 in the brain. Several forms of tau phosphorylated at threonine 181 have been shown to contribute to synapse degeneration in Alzheimer's disease, which correlates closely with cognitive decline in this form of dementia, but to date, there have not been investigations of whether tau phosphorylated at threonine 181 is found in synapses in Alzheimer's disease or healthy ageing brain. It was also previously unclear whether tau phosphorylated at threonine 181 accumulated in dystrophic neurites around plaques, which could contribute to tau leakage to the periphery due to impaired membrane integrity in dystrophies. Brain homogenate and biochemically enriched synaptic fractions were examined with western blot to examine tau phosphorylated at threonine 181 levels between groups (n = 10-12 per group), and synaptic and astrocytic localization of tau phosphorylated at threonine 181 were examined using array tomography (n = 6-15 per group), and localization of tau phosphorylated at threonine 181 in plaque-associated dystrophic neurites with associated gliosis were examined with standard immunofluorescence (n = 8-9 per group). Elevated baseline plasma tau phosphorylated at threonine 181, neurofilament light and fibrillary acidic protein predicted steeper general cognitive decline during ageing. Further, increasing tau phosphorylated at threonine 181 over time predicted general cognitive decline in females only. Change in plasma tau phosphorylated at threonine 181 remained a significant predictor of g factor decline when taking into account Alzheimer's disease polygenic risk score, indicating that the increase of blood tau phosphorylated at threonine 181 in this cohort was not only due to incipient Alzheimer's disease. Tau phosphorylated at threonine 181 was observed in synapses and astrocytes in both healthy ageing and Alzheimer's disease brain. We observed that a significantly higher proportion of synapses contain tau phosphorylated at threonine 181 in Alzheimer's disease relative to aged controls. Aged controls with pre-morbid lifetime cognitive resilience had significantly more tau phosphorylated at threonine 181 in fibrillary acidic protein-positive astrocytes than those with pre-morbid lifetime cognitive decline. Further, tau phosphorylated at threonine 181 was found in dystrophic neurites around plaques and in some neurofibrillary tangles. The presence of tau phosphorylated at threonine 181 in plaque-associated dystrophies may be a source of leakage of tau out of neurons that eventually enters the blood. Together, these data indicate that plasma tau phosphorylated at threonine 181, neurofilament light and fibrillary acidic protein may be useful biomarkers of age-related cognitive decline, and that efficient clearance of tau phosphorylated at threonine 181 by astrocytes may promote cognitive resilience.
Collapse
Affiliation(s)
- Tyler S Saunders
- UK Dementia Research Institute and Centre for Discovery Brain Sciences at the University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Edinburgh Dementia Prevention & Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Francesca E Pozzolo
- UK Dementia Research Institute and Centre for Discovery Brain Sciences at the University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Amanda Heslegrave
- United Kingdom UK Dementia Research Institute at University College London, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Declan King
- UK Dementia Research Institute and Centre for Discovery Brain Sciences at the University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Robert I McGeachan
- UK Dementia Research Institute and Centre for Discovery Brain Sciences at the University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Maxwell P Spires-Jones
- UK Dementia Research Institute and Centre for Discovery Brain Sciences at the University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Sarah E Harris
- Lothian Birth Cohort studies, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9AD, UK
| | - Craig Ritchie
- Edinburgh Dementia Prevention & Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Graciela Muniz-Terrera
- Edinburgh Dementia Prevention & Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH4 2XU, UK
- Department of Social Medicine, Ohio University, Athens, Ohio 45701, USA
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago 3485, Chile
| | - Ian J Deary
- Lothian Birth Cohort studies, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9AD, UK
| | - Simon R Cox
- Lothian Birth Cohort studies, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9AD, UK
| | - Henrik Zetterberg
- United Kingdom UK Dementia Research Institute at University College London, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, S-431 80 Molndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80 Molndal, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Tara L Spires-Jones
- UK Dementia Research Institute and Centre for Discovery Brain Sciences at the University of Edinburgh, Edinburgh, EH8 9JZ, UK
| |
Collapse
|
13
|
Yu X, Shao K, Wan K, Li T, Li Y, Zhu X, Han Y. Progress in blood biomarkers of subjective cognitive decline in preclinical Alzheimer's disease. Chin Med J (Engl) 2023; 136:505-521. [PMID: 36914945 PMCID: PMC10106168 DOI: 10.1097/cm9.0000000000002566] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Indexed: 03/15/2023] Open
Abstract
ABSTRACT Alzheimer's disease (AD) is a neurodegenerative disease that gradually impairs cognitive functions. Recently, there has been a conceptual shift toward AD to view the disease as a continuum. Since AD is currently incurable, effective intervention to delay or prevent pathological cognitive decline may best target the early stages of symptomatic disease, such as subjective cognitive decline (SCD), in which cognitive function remains relatively intact. Diagnostic methods for identifying AD, such as cerebrospinal fluid biomarkers and positron emission tomography, are invasive and expensive. Therefore, it is imperative to develop blood biomarkers that are sensitive, less invasive, easier to access, and more cost effective for AD diagnosis. This review aimed to summarize the current data on whether individuals with SCD differ reliably and effectively in subjective and objective performances compared to cognitively normal elderly individuals, and to find one or more convenient and accessible blood biomarkers so that researchers can identify SCD patients with preclinical AD in the population as soon as possible. Owing to the heterogeneity and complicated pathogenesis of AD, it is difficult to make reliable diagnoses using only a single blood marker. This review provides an overview of the progress achieved to date with the use of SCD blood biomarkers in patients with preclinical AD, highlighting the key areas of application and current challenges.
Collapse
Affiliation(s)
- Xianfeng Yu
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Kai Shao
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Ke Wan
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Taoran Li
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Yuxia Li
- Department of Neurology, Tangshan Central Hospital, Tangshan, Hebei 063000, China
| | - Xiaoqun Zhu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
- School of Biomedical Engineering, Hainan University, Haikou, Hainan 570228, China
- Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Clinical Research Center for Geriatric Diseases, Beijing 100053, China
| |
Collapse
|
14
|
Behzad M, Zirak N, Madani GH, Baidoo L, Rezaei A, Karbasi S, Sadeghi M, Shafie M, Mayeli M, Alzheimer's Disease Neuroimaging Initiative. CSF-Targeted Proteomics Indicate Amyloid-Beta Ratios in Patients with Alzheimer's Dementia Spectrum. Int J Alzheimers Dis 2023; 2023:5336273. [PMID: 36793451 PMCID: PMC9925239 DOI: 10.1155/2023/5336273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 12/01/2022] [Accepted: 12/07/2022] [Indexed: 02/08/2023] Open
Abstract
Background According to recent studies, amyloid-β (Aβ) isoforms as cerebrospinal fluid (CSF) biomarkers have remarkable predictive value for cognitive decline in the early stages of Alzheimer's disease (AD). Herein, we aimed to investigate the correlations between several targeted proteomics in CSF samples with Aβ ratios and cognitive scores in patients in AD spectrum to search for potential early diagnostic utility. Methods A total of 719 participants were found eligible for inclusion. Patients were then categorized into cognitively normal (CN), mild cognitive impairment (MCI), and AD and underwent an assessment of Aβ and proteomics. Clinical Dementia Rating (CDR), Alzheimer's Disease Assessment Scale (ADAS), and Mini Mental State Exam (MMSE) were used for further cognitive assessment. The Aβ42, Aβ42/Aβ40, and Aβ42/38 ratios were considered as means of comparison to identify those peptides corresponding significantly to these established biomarkers and cognitive scores. The diagnostic utility of the IASNTQSR, VAELEDEK, VVSSIEQK, GDSVVYGLR, EPVAGDAVPGPK, and QETLPSK was assessed. Results All investigated peptides corresponded significantly to Aβ42 in controls. In those with MCI, VAELEDEK and EPVAGDAVPGPK were significantly correlated with Aβ42 (p value < 0.001). Additionally, IASNTQSR, VVSSIEQK, GDSVVYGLR, and QETLPSK were significantly correlated with Aβ42/Aβ40 and Aβ42/38 (p value < 0.001) in this group. This group of peptides similarly corresponded to Aβ ratios in those with AD. Eventually, IASNTQSR, VAELEDEK, and VVSSIEQK were significantly associated with CDR, ADAS-11, and ADAS-13, particularly in MCI group. Conclusion Our research suggests potential early diagnostic and prognostic utilities for certain peptides extracted from CSF-targeted proteomics research. The ethical approval of ADNI is available at ClinicalTrials.gov with Identifier: NCT00106899.
Collapse
Affiliation(s)
- Maryam Behzad
- NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Chemistery, University of Tehran, Iran
| | - Negin Zirak
- NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Educational Science and Psychology, University of Tabriz, Tabriz, Iran
| | - Ghazal Hamidi Madani
- NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Biology, Faculty of Sciences, University of Guilan, Iran
| | - Linda Baidoo
- NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Rezaei
- NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Karbasi
- NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Sadeghi
- NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahan Shafie
- NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Mayeli
- NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
15
|
Pascual-Lucas M, Allué JA, Sarasa L, Fandos N, Castillo S, Terencio J, Sarasa M, Tartari JP, Sanabria Á, Tárraga L, Ruíz A, Marquié M, Seo SW, Jang H, Boada M. Clinical performance of an antibody-free assay for plasma Aβ42/Aβ40 to detect early alterations of Alzheimer's disease in individuals with subjective cognitive decline. Alzheimers Res Ther 2023; 15:2. [PMID: 36604729 PMCID: PMC9814201 DOI: 10.1186/s13195-022-01143-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/14/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Accessible and cost-effective diagnostic tools are urgently needed to accurately quantify blood biomarkers to support early diagnosis of Alzheimer's disease (AD). In this study, we investigated the ability of plasma amyloid-beta (Aβ)42/Aβ40 ratio measured by an antibody-free mass-spectrometric (MS) method, ABtest-MS, to detect early pathological changes of AD. METHODS This cohort study included data from the baseline and 2-year follow-up visits from the Fundació ACE Healthy Brain Initiative (FACEHBI) study. Plasma Aβ42/Aβ40 was measured with ABtest-MS and compared to 18F-Florbetaben PET as the reference standard (cutoff for early amyloid deposition of 13.5 centiloids). Cross-validation was performed in an independent DPUK-Korean cohort. Additionally, associations of plasma Aβ42/Aβ40 with episodic memory performance and brain atrophy were assessed. RESULTS The FACEHBI cohort at baseline included 200 healthy individuals with subjective cognitive decline (SCD), of which 36 (18%) were Aβ-PET positive. Plasma Aβ42/Aβ40 levels were significantly lower in Aβ-PET positive individuals (median [interquartile range, IQR], 0.215 [0.203-0.236]) versus Aβ-PET negative subjects (median [IQR], 0.261 [0.244-0.279]) (P < .001). Plasma Aβ42/Aβ40 was significantly correlated with Aβ-PET levels (rho = -0.390; P < .001) and identified Aβ-PET status with an area under the receiver operating characteristic curve (AUC) of 0.87 (95% confidence interval [CI], 0.80-0.93). A cutoff for the Aβ42/Aβ40 ratio of 0.241 (maximum Youden index) yielded a sensitivity of 86.1% and a specificity of 80.5%. These findings were cross-validated in an independent DPUK-Korean cohort (AUC 0.86 [95% CI 0.77-0.95]). Lower plasma Aβ42/Aβ40 ratio was associated with worse episodic memory performance and increased brain atrophy. Plasma Aβ42/Aβ40 at baseline predicted clinical conversion to mild cognitive impairment and longitudinal changes in amyloid deposition and brain atrophy at 2-year follow-up. CONCLUSIONS This study suggests that plasma Aβ42/Aβ40, as determined by this MS-based assay, has potential value as an accurate and cost-effective tool to identify individuals in the earliest stages of AD, supporting its implementation in clinical trials, preventative strategies and clinical practice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Juan Pablo Tartari
- grid.410675.10000 0001 2325 3084Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
| | - Ángela Sanabria
- grid.410675.10000 0001 2325 3084Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain ,grid.418264.d0000 0004 1762 4012CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Lluís Tárraga
- grid.410675.10000 0001 2325 3084Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain ,grid.418264.d0000 0004 1762 4012CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Agustín Ruíz
- grid.410675.10000 0001 2325 3084Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain ,grid.418264.d0000 0004 1762 4012CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Marta Marquié
- grid.410675.10000 0001 2325 3084Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain ,grid.418264.d0000 0004 1762 4012CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Sang Won Seo
- grid.264381.a0000 0001 2181 989XDepartment of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hyemin Jang
- grid.264381.a0000 0001 2181 989XDepartment of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Mercè Boada
- grid.410675.10000 0001 2325 3084Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain ,grid.418264.d0000 0004 1762 4012CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | | |
Collapse
|
16
|
Stark J, Hiersche KJ, Yu JC, Hasselbach AN, Abdi H, Hayes SM. Partial Least Squares Regression Analysis of Alzheimer's Disease Biomarkers, Modifiable Health Variables, and Cognitive Change in Older Adults with Mild Cognitive Impairment. J Alzheimers Dis 2023; 93:633-651. [PMID: 37066909 PMCID: PMC10999056 DOI: 10.3233/jad-221084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
BACKGROUND Prior work has shown that certain modifiable health, Alzheimer's disease (AD) biomarker, and demographic variables are associated with cognitive performance. However, less is known about the relative importance of these different domains of variables in predicting longitudinal change in cognition. OBJECTIVE Identify novel relationships between modifiable physical and health variables, AD biomarkers, and slope of cognitive change over two years in a cohort of older adults with mild cognitive impairment (MCI). METHODS Metrics of cardiometabolic risk, stress, inflammation, neurotrophic/growth factors, and AD pathology were assessed in 123 older adults with MCI at baseline from the Alzheimer's Disease Neuroimaging Initiative (mean age = 73.9; SD = 7.6; mean education = 16.0; SD = 3.0). Partial least squares regression (PLSR)-a multivariate method which creates components that best predict an outcome-was used to identify whether these physiological variables were important in predicting slope of change in episodic memory or executive function over two years. RESULTS At two-year follow-up, the two PLSR models predicted, respectively, 20.0% and 19.6% of the variance in change in episodic memory and executive function. Baseline levels of AD biomarkers were important in predicting change in both episodic memory and executive function. Baseline education and neurotrophic/growth factors were important in predicting change in episodic memory, whereas cardiometabolic variables such as blood pressure and cholesterol were important in predicting change in executive function. CONCLUSION These data-driven analyses highlight the impact of AD biomarkers on cognitive change and further clarify potential domain specific relationships with predictors of cognitive change.
Collapse
Affiliation(s)
- Jessica Stark
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Kelly J Hiersche
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Ju-Chi Yu
- Centre for Addiction and Mental Health, Toronto, Canada
| | | | - Hervé Abdi
- Department of Psychology, The University of Texas at Dallas, Dallas, TX, USA
| | - Scott M Hayes
- Department of Psychology, The Ohio State University, Columbus, OH, USA
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
17
|
Sanchez-Sanchez JL, Giudici KV, Guyonnet S, Delrieu J, Li Y, Bateman RJ, Parini A, Vellas B, de Souto Barreto P, Vellas B, Guyonnet S, Carrié I, Brigitte L, Faisant C, Lala F, Delrieu J, Villars H, Combrouze E, Badufle C, Zueras A, Andrieu S, Cantet C, Morin C, Van Kan GA, Dupuy C, Rolland Y, Caillaud C, Ousset PJ, Lala F, Willis S, Belleville S, Gilbert B, Fontaine F, Dartigues JF, Marcet I, Delva F, Foubert A, Cerda S, Marie-Noëlle-Cuffi, Costes C, Rouaud O, Manckoundia P, Quipourt V, Marilier S, Franon E, Bories L, Pader ML, Basset MF, Lapoujade B, Faure V, Tong MLY, Malick-Loiseau C, Cazaban-Campistron E, Desclaux F, Blatge C, Dantoine T, Laubarie-Mouret C, Saulnier I, Clément JP, Picat MA, Bernard-Bourzeix L, Willebois S, Désormais I, Cardinaud N, Bonnefoy M, Livet P, Rebaudet P, Gédéon C, Burdet C, Terracol F, Pesce A, Roth S, Chaillou S, Louchart S, Sudres K, Lebrun N, Barro-Belaygues N, Touchon J, Bennys K, Gabelle A, Romano A, Touati L, Marelli C, Pays C, Robert P, Le Duff F, Gervais C, Gonfrier S, Gasnier Y, Bordes S, Begorre D, Carpuat C, Khales K, Lefebvre JF, El Idrissi SM, Skolil P, Salles JP, Dufouil C, Lehéricy S, Chupin M, et alSanchez-Sanchez JL, Giudici KV, Guyonnet S, Delrieu J, Li Y, Bateman RJ, Parini A, Vellas B, de Souto Barreto P, Vellas B, Guyonnet S, Carrié I, Brigitte L, Faisant C, Lala F, Delrieu J, Villars H, Combrouze E, Badufle C, Zueras A, Andrieu S, Cantet C, Morin C, Van Kan GA, Dupuy C, Rolland Y, Caillaud C, Ousset PJ, Lala F, Willis S, Belleville S, Gilbert B, Fontaine F, Dartigues JF, Marcet I, Delva F, Foubert A, Cerda S, Marie-Noëlle-Cuffi, Costes C, Rouaud O, Manckoundia P, Quipourt V, Marilier S, Franon E, Bories L, Pader ML, Basset MF, Lapoujade B, Faure V, Tong MLY, Malick-Loiseau C, Cazaban-Campistron E, Desclaux F, Blatge C, Dantoine T, Laubarie-Mouret C, Saulnier I, Clément JP, Picat MA, Bernard-Bourzeix L, Willebois S, Désormais I, Cardinaud N, Bonnefoy M, Livet P, Rebaudet P, Gédéon C, Burdet C, Terracol F, Pesce A, Roth S, Chaillou S, Louchart S, Sudres K, Lebrun N, Barro-Belaygues N, Touchon J, Bennys K, Gabelle A, Romano A, Touati L, Marelli C, Pays C, Robert P, Le Duff F, Gervais C, Gonfrier S, Gasnier Y, Bordes S, Begorre D, Carpuat C, Khales K, Lefebvre JF, El Idrissi SM, Skolil P, Salles JP, Dufouil C, Lehéricy S, Chupin M, Mangin JF, Bouhayia A, Allard M, Ricolfi F, Dubois D, Martel MPB, Cotton F, Bonafé A, Chanalet S, Hugon F, Bonneville F, Cognard C, Chollet F, Payoux P, Voisin T, Peiffer S, Hitzel A, Zanca M, Monteil J, Darcourt J, Molinier L, Derumeaux H, Costa N, Perret B, Vinel C, Caspar-Bauguil S, Olivier-Abbal P, Coley N. Plasma MCP-1 and changes on cognitive function in community-dwelling older adults. Alzheimers Res Ther 2022; 14:5. [PMID: 34996522 PMCID: PMC8742409 DOI: 10.1186/s13195-021-00940-2] [Show More Authors] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/16/2021] [Indexed: 12/24/2022]
Abstract
Background Monocyte Chemoattractant Protein-1 (MCP-1), a glial-derived chemokine, mediates neuroinflammation and may regulate memory outcomes among older adults. We aimed to explore the associations of plasma MCP-1 levels (alone and in combination with β-amyloid deposition—Aβ42/40) with overall and domain-specific cognitive evolution among older adults. Methods Secondary analyses including 1097 subjects (mean age = 75.3 years ± 4.4; 63.8% women) from the Multidomain Alzheimer Preventive Trial (MAPT). MCP-1 (higher is worse) and Aβ42/40 (lower is worse) were measured in plasma collected at year 1. MCP-1 in continuous and as a dichotomy (values in the highest quartile (MCP-1+)) were used, as well as a dichotomy of Aβ42/40. Outcomes were measured annually over 4 years and included the following: cognitive composite z-score (CCS), the Mini-Mental State Examination (MMSE), and Clinical Dementia Rating (CDR) sum of boxes (overall cognitive function); composite executive function z-score, composite attention z-score, Free and Cued Selective Reminding Test (FCSRT - memory). Results Plasma MCP-1 as a continuous variable was associated with the worsening of episodic memory over 4 years of follow-up, specifically in measures of free and cued delayed recall. MCP-1+ was associated with worse evolution in the CCS (4-year between-group difference: β = −0.14, 95%CI = −0.26, −0.02) and the CDR sum of boxes (2-year: β = 0.19, 95%CI = 0.06, 0.32). In domain-specific analyses, MCP-1+ was associated with declines in the FCSRT delayed recall sub-domains. In the presence of low Aβ42/40, MCP-1+ was not associated with greater declines in cognitive functions. The interaction with continuous biomarker values Aβ42/40× MCP-1 × time was significant in models with CDR sum of boxes and FCSRT DTR as dependent variables. Conclusions Baseline plasma MCP-1 levels were associated with longitudinal declines in overall cognitive and episodic memory performance in older adults over a 4-year follow-up. How plasma MCP-1 interacts with Aβ42/40 to determine cognitive decline at different stages of cognitive decline/dementia should be clarified by further research. The MCP-1 association on cognitive decline was strongest in those with amyloid plaques, as measured by blood plasma Aβ42/40. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00940-2.
Collapse
|
18
|
Aschenbrenner AJ, Li Y, Henson RL, Volluz K, Hassenstab J, Verghese P, West T, Meyer MR, Kirmess KM, Fagan AM, Xiong C, Holtzman D, Morris JC, Bateman RJ, Schindler SE. Comparison of plasma and CSF biomarkers in predicting cognitive decline. Ann Clin Transl Neurol 2022; 9:1739-1751. [PMID: 36183195 PMCID: PMC9639639 DOI: 10.1002/acn3.51670] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/07/2022] [Indexed: 11/07/2022] Open
Abstract
OBJECTIVES Concentrations of amyloid-β peptides (Aβ42/Aβ40) and neurofilament light (NfL) can be measured in plasma or cerebrospinal fluid (CSF) and are associated with Alzheimer's disease brain pathology and cognitive impairment. This study directly compared plasma and CSF measures of Aβ42/Aβ40 and NfL as predictors of cognitive decline. METHODS Participants were 65 years or older and cognitively normal at baseline with at least one follow-up cognitive assessment. Analytes were measured with the following types of assays: plasma Aβ42/Aβ40, immunoprecipitation-mass spectrometry; plasma NfL, Simoa; CSF Aβ42/Aβ40, automated immunoassay; CSF NfL plate-based immunoassay. Mixed effects models evaluated the global cognitive composite score over a maximum of 6 years as predicted by the fluid biomarkers. RESULTS Analyses included 371 cognitively normal participants, aged 72.7 ± 5.2 years (mean ± standard deviation) with an average length of follow-up of 3.9 ± 1.6 years. Standardized concentrations of biomarkers were associated with annualized cognitive change: plasma Aβ42/Aβ40, 0.014 standard deviations (95% confidence intervals 0.002 to 0.026); CSF Aβ42/Aβ40, 0.020 (0.008 to 0.032); plasma Nfl, -0.018 (-0.030 to -0.005); and CSF NfL, -0.024 (-0.036 to -0.012). Power analyses estimated that 266 individuals in each treatment arm would be needed to detect a 50% slowing of decline if identified by abnormal plasma measures versus 229 for CSF measures. INTERPRETATION Both plasma and CSF measures of Aβ42/Aβ40 and NfL predicted cognitive decline. A clinical trial that enrolled individuals based on abnormal plasma Aβ42/Aβ40 and NfL levels would require only a marginally larger cohort than if CSF measures were used.
Collapse
Affiliation(s)
- Andrew J. Aschenbrenner
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
| | - Yan Li
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
- Division of BiostatisticsWashington University School of MedicineSt. LouisMOUSA
| | - Rachel L. Henson
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
| | - Katherine Volluz
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
| | - Jason Hassenstab
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
| | | | | | | | | | - Anne M. Fagan
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
- Hope Center for Neurological DisordersWashington University School of MedicineSt. LouisMOUSA
| | - Chengjie Xiong
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
- Division of BiostatisticsWashington University School of MedicineSt. LouisMOUSA
| | - David Holtzman
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
- Hope Center for Neurological DisordersWashington University School of MedicineSt. LouisMOUSA
| | - John C. Morris
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
- Hope Center for Neurological DisordersWashington University School of MedicineSt. LouisMOUSA
| | - Randall J. Bateman
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
- Hope Center for Neurological DisordersWashington University School of MedicineSt. LouisMOUSA
| | - Suzanne E. Schindler
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
| |
Collapse
|
19
|
Huang Y, Li Y, Xie F, Guo Q. Associations of plasma phosphorylated tau181 and neurofilament light chain with brain amyloid burden and cognition in objectively defined subtle cognitive decline patients. CNS Neurosci Ther 2022; 28:2195-2205. [PMID: 36074638 PMCID: PMC9627371 DOI: 10.1111/cns.13962] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/05/2022] [Accepted: 08/15/2022] [Indexed: 02/06/2023] Open
Abstract
AIMS There is increasing evidence that plasma biomarkers are specific biomarkers for Alzheimer's disease (AD) pathology, but their potential utility in Obj-SCD (objectively defined subtle cognitive decline) remains unclear. METHODS A total of 234 subjects, including 65 with brain amyloid beta (Aβ) negative normal cognition (Aβ- NC), 58 with Aβ-positive NC (Aβ+ NC), 63 with Aβ- Obj-SCD, and 48 with Aβ+ Obj-SCD were enrolled. Plasma Aβ42, Aβ40, Aβ42/Aβ40 ratio, phosphorylated tau181 (p-tau181), neurofilament light chain (NfL), and total tau (T-tau) were measured using Simoa assays. Logistic and linear regression analyses were used to examine the relationship between plasma biomarkers and brain amyloid, cognition, and imaging measures adjusting for age, sex, education, APOE ε4 status, and vascular risk scores. Receiver operating characteristics were used to evaluate the discriminative validity of biomarkers. RESULTS After adjustment, only plasma p-tau181 and NfL were significantly elevated in Aβ+ Obj-SCD participants compared to Aβ- NC group. Elevated p-tau181 was associated with brain amyloid accumulation, worse cognitive performance (visual episodic memory, executive function, and visuospatial function), and hippocampal atrophy. These associations mainly occurred in Aβ+ individuals. In contrast, higher NfL was correlated with brain amyloid burden and verbal memory decline. These associations predominantly occurred in Aβ- individuals. The adjusted diagnostic model combining p-tau181 and NfL levels showed the best performance in identifying Aβ+ Obj-SCD from Aβ- NC [area under the curve (AUC) = 0.814], which did not differ from the adjusted p-tau181 model (AUC = 0.763). CONCLUSIONS Our findings highlight that plasma p-tau181, alone or combined with NfL, contributes to identifying high-risk AD populations.
Collapse
Affiliation(s)
- Yanlu Huang
- Department of GerontologyShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Yuehua Li
- Department of RadiologyShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Fang Xie
- PET Center, Huashan HospitalFudan UniversityShanghaiChina
| | - Qihao Guo
- Department of GerontologyShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| |
Collapse
|
20
|
Morrison C, Dadar M, Villeneuve S, Collins DL. White matter lesions may be an early marker for age-related cognitive decline. Neuroimage Clin 2022; 35:103096. [PMID: 35764028 PMCID: PMC9241138 DOI: 10.1016/j.nicl.2022.103096] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/16/2022] [Accepted: 06/19/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Research suggests that cerebral small vessel disease (CSVD), amyloid, and pTau contribute to age-related cognitive decline. It remains unknown how these factors relate to one another and how they jointly contribute to cognitive decline in normal aging. This project examines the association between these factors and their relationship to cognitive decline in cognitively unimpaired older adults without subjective cognitive decline. METHODS A total of 230 subjects with cerebrospinal fluid (CSF) Aß42, CSF pTau181, white matter lesions (WMLs) used as a proxy of CSVD, and cognitive scores from the Alzheimer's Disease Neuroimaging Initiative were included. Associations between each factor and cognitive score were investigated using regression models. Furthermore, relationships between the three pathologies were also examined using regression models. RESULTS At baseline, there was an inverse association between WML load and Aß42 (t = -4.20, p <.001). There was no association between WML load and pTau (t = 0.32, p = 0.75), nor with Aß42 and pTau (t = 0.51, p =.61). Correcting for age, sex and education, baseline WML load was associated with baseline ADAS-13 scores (t = 2.59, p =.01) and lower follow-up executive functioning (t = -2.84, p =.005). Baseline Aß42 was associated with executive function at baseline (t = 3.58, p<.004) but not at follow-up (t = 1.05, p = 0.30), nor with ADAS-13 at baseline (t = -0.24, p = 0.81) or follow-up (t = 0.09, p = 0.93). Finally, baseline pTau was not associated with any cognitive measure at baseline or follow-up. CONCLUSION Both baseline Aß42 and WML load are associated with some baseline cognition scores, but only baseline WML load is associated with follow-up executive functioning. This finding suggests that WMLs may be one of the earliest clinical manifestations that contributes to future cognitive decline in cognitively healthy older adults. Given that healthy older adults with WMLs exhibit declines in cognitive functioning, they may be less resilient to future pathology increasing their risk for cognitive impairment due to dementia than those without WMLs.
Collapse
Affiliation(s)
- Cassandra Morrison
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, H3A 2B4 Montreal, Quebec, Canada.
| | - Mahsa Dadar
- Department of Psychiatry, McGill University, H3A 1A1 Montreal, Quebec, Canada; Douglas Mental Health University Institute, Studies on Prevention of Alzheimer's Disease (StoP-AD) Centre, H4H 1R3 Montreal, Quebec, Canada
| | - Sylvia Villeneuve
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, H3A 2B4 Montreal, Quebec, Canada; Department of Psychiatry, McGill University, H3A 1A1 Montreal, Quebec, Canada; Douglas Mental Health University Institute, Studies on Prevention of Alzheimer's Disease (StoP-AD) Centre, H4H 1R3 Montreal, Quebec, Canada
| | - D Louis Collins
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, H3A 2B4 Montreal, Quebec, Canada
| |
Collapse
|
21
|
Fernandez-Alvarez M, Atienza M, Zallo F, Matute C, Capetillo-Zarate E, Cantero JL. Linking Plasma Amyloid Beta and Neurofilament Light Chain to Intracortical Myelin Content in Cognitively Normal Older Adults. Front Aging Neurosci 2022; 14:896848. [PMID: 35783126 PMCID: PMC9247578 DOI: 10.3389/fnagi.2022.896848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/23/2022] [Indexed: 11/29/2022] Open
Abstract
Evidence suggests that lightly myelinated cortical regions are vulnerable to aging and Alzheimer’s disease (AD). However, it remains unknown whether plasma markers of amyloid and neurodegeneration are related to deficits in intracortical myelin content, and whether this relationship, in turn, is associated with altered patterns of resting-state functional connectivity (rs-FC). To shed light into these questions, plasma levels of amyloid-β fragment 1–42 (Aβ1–42) and neurofilament light chain (NfL) were measured using ultra-sensitive single-molecule array (Simoa) assays, and the intracortical myelin content was estimated with the ratio T1-weigthed/T2-weighted (T1w/T2w) in 133 cognitively normal older adults. We assessed: (i) whether plasma Aβ1–42 and/or NfL levels were associated with intracortical myelin content at different cortical depths and (ii) whether cortical regions showing myelin reductions also exhibited altered rs-FC patterns. Surface-based multiple regression analyses revealed that lower plasma Aβ1–42 and higher plasma NfL were associated with lower myelin content in temporo-parietal-occipital regions and the insular cortex, respectively. Whereas the association with Aβ1–42 decreased with depth, the NfL-myelin relationship was most evident in the innermost layer. Older individuals with higher plasma NfL levels also exhibited altered rs-FC between the insula and medial orbitofrontal cortex. Together, these findings establish a link between plasma markers of amyloid/neurodegeneration and intracortical myelin content in cognitively normal older adults, and support the role of plasma NfL in boosting aberrant FC patterns of the insular cortex, a central brain hub highly vulnerable to aging and neurodegeneration.
Collapse
Affiliation(s)
- Marina Fernandez-Alvarez
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Seville, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Mercedes Atienza
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Seville, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Fatima Zallo
- Departamento de Neurociencias, Achucarro Basque Center for Neuroscience, Universidad del País Vasco, Leioa, Spain
| | - Carlos Matute
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Departamento de Neurociencias, Achucarro Basque Center for Neuroscience, Universidad del País Vasco, Leioa, Spain
| | - Estibaliz Capetillo-Zarate
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Departamento de Neurociencias, Achucarro Basque Center for Neuroscience, Universidad del País Vasco, Leioa, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Jose L. Cantero
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Seville, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- *Correspondence: Jose L. Cantero,
| |
Collapse
|
22
|
Lu WH, Giudici KV, Morley JE, Guyonnet S, Parini A, Aggarwal G, Nguyen AD, Li Y, Bateman RJ, Vellas B, de Souto Barreto P. Investigating the combination of plasma amyloid-beta and geroscience biomarkers on the incidence of clinically meaningful cognitive decline in older adults. GeroScience 2022; 44:1489-1503. [PMID: 35445358 PMCID: PMC9213609 DOI: 10.1007/s11357-022-00554-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 03/22/2022] [Indexed: 11/04/2022] Open
Abstract
We investigated combining a core AD neuropathology measure (plasma amyloid-beta [Aβ] 42/40) with five plasma markers of inflammation, cellular stress, and neurodegeneration to predict cognitive decline. Among 401 participants free of dementia (median [IQR] age, 76 [73-80] years) from the Multidomain Alzheimer Preventive Trial (MAPT), 28 (7.0%) participants developed dementia, and 137 (34.2%) had worsening of clinical dementia rating (CDR) scale over 4 years. In the models utilizing plasma Aβ alone, a tenfold increased risk of incident dementia (nonsignificant) and a fivefold increased risk of worsening CDR were observed as each nature log unit increased in plasma Aβ levels. Models incorporating Aβ plus multiple plasma biomarkers performed similarly to models included Aβ alone in predicting dementia and CDR progression. However, improving Aβ model performance for composite cognitive score (CCS) decline, a proxy of dementia, was observed after including plasma monocyte chemoattractant protein 1 (MCP1) and growth differentiation factor 15 (GDF15) as covariates. Participants with abnormal Aβ, GDF15, and MCP1 presented higher CCS decline (worsening cognitive function) compared to their normal-biomarker counterparts (adjusted β [95% CI], - 0.21 [- 0.35 to - 0.06], p = 0.005). In conclusion, our study found limited added values of multi-biomarkers beyond the basic Aβ models for predicting clinically meaningful cognitive decline among non-demented older adults. However, a combined assessment of inflammatory and cellular stress status with Aβ pathology through measuring plasma biomarkers may improve the evaluation of cognitive performance.
Collapse
Affiliation(s)
- Wan-Hsuan Lu
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), 31000, Toulouse, France
- Inserm CERPOP - UMR1295, University of Toulouse III, 31000, Toulouse, France
| | - Kelly Virecoulon Giudici
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), 31000, Toulouse, France
| | - John E Morley
- Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Sophie Guyonnet
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), 31000, Toulouse, France
- Inserm CERPOP - UMR1295, University of Toulouse III, 31000, Toulouse, France
| | - Angelo Parini
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Inserm UMR 1048, University of Toulouse, 31400, Toulouse, France
| | - Geetika Aggarwal
- Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, MO, USA
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, USA
| | - Andrew D Nguyen
- Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, MO, USA
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, USA
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Bruno Vellas
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), 31000, Toulouse, France
- Inserm CERPOP - UMR1295, University of Toulouse III, 31000, Toulouse, France
| | - Philipe de Souto Barreto
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), 31000, Toulouse, France
- Inserm CERPOP - UMR1295, University of Toulouse III, 31000, Toulouse, France
| |
Collapse
|
23
|
Bhagavati S. Commentary: Diagnostic Accuracy of Blood-Based Biomarker Panels: A Systematic Review. Front Aging Neurosci 2022; 14:895398. [PMID: 35572139 PMCID: PMC9099371 DOI: 10.3389/fnagi.2022.895398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 04/13/2022] [Indexed: 11/30/2022] Open
|
24
|
Wang J, Gao L, Liu J, Dang L, Wei S, Hu N, Gao Y, Peng W, Shang S, Huo K, Wang J, Qu Q. The Association of Plasma Amyloid-β and Cognitive Decline in Cognitively Unimpaired Population. Clin Interv Aging 2022; 17:555-565. [PMID: 35480964 PMCID: PMC9035463 DOI: 10.2147/cia.s357994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022] Open
Abstract
Purpose This study investigates the relationship between baseline plasma Aβ and cognitive decline during follow-up in cognitively unimpaired population. Materials and Methods Cognitively unimpaired population was selected from people who lived in the suburbs of Xi’an, China. The levels of plasma Aβ1-42 and Aβ1-40 were tested using commercial enzyme-linked immunosorbent assay (ELISA). The mini-mental state examination (MMSE) and neuropsychological battery were used to assess cognition. Two years later, MMSE was tested again, and significant cognitive decline was defined as a decrease in MMSE scores ≥5 points. Logistic regression analysis was performed to analyze the relationship between baseline plasma Aβ and cognitive change during the two-year follow-up. Results A total of 1144 participants completed the study, among whom 59 subjects (5.2%) presented significant cognitive decline. The high plasma Aβ1-42 level group had more significant cognitive decline (P = 0.023). Multivariable logistic regression analysis showed that significant cognitive decline was associated with the high levels of baseline plasma Aβ1-42 (OR = 1.043, 95% CI: 1.005–1.083, P = 0.026). However, significant cognitive decline was not associated with baseline plasma Aβ1-40 levels and Aβ1-42 /Aβ1-40 ratio. Conclusion Population with high level of baseline plasma Aβ1-42 manifested significant cognitive decline over 2 years; however, further investigation on the dynamics of plasma Aβ and long-term follow-up are needed.
Collapse
Affiliation(s)
- Jin Wang
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Ling Gao
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Jie Liu
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Liangjun Dang
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Shan Wei
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Ningwei Hu
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Yao Gao
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Wei Peng
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Suhang Shang
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Kang Huo
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Jingyi Wang
- Huyi Hospital of Traditional Chinese Medicine, Xi’an, People’s Republic of China
| | - Qiumin Qu
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
- Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
- Correspondence: Qiumin Qu, Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an, 710061, People’s Republic of China, Tel/Fax +86 29 8532 4083, Email
| |
Collapse
|
25
|
APOE-ε4 modulates the association among plasma Aβ 42/Aβ 40, vascular diseases, neurodegeneration and cognitive decline in non-demented elderly adults. Transl Psychiatry 2022; 12:128. [PMID: 35351867 PMCID: PMC8964707 DOI: 10.1038/s41398-022-01899-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/03/2022] [Accepted: 03/11/2022] [Indexed: 01/18/2023] Open
Abstract
Including apolipoprotein E-ε4 (APOE-ε4) status and older age into consideration may increase the accuracy of plasma Aβ42/Aβ40 detecting Aβ+ individuals, but the rationale behind this remains to be fully understood. Besides, both Aβ pathology and vascular diseases are related to neurodegeneration and cognitive decline, but it is still not fully understood how APOE-ε4 modulates these relationships. In this study, we examined 241 non-demented Alzheimer's Disease Neuroimaging Initiative participants to investigate the associations among age, white matter hyperintensities (WMH), hypertension, hyperlipidemia, body mass index (BMI), plasma Aβ42/Aβ40 measured by liquid chromatography tandem mass spectrometry, and 18F-florbetapir Aβ PET as well as their prediction of longitudinal adjusted hippocampal volume (aHCV) and cognition in APOE-ε4 carriers and non-carriers. We found older age predicted faster WMH increase (p = 0.024) and cortical Aβ accumulation (p = 0.043) in APOE-ε4 non-carriers only, whereas lower plasma Aβ42/Aβ40 predicted faster cortical Aβ accumulation (p < 0.018) regardless of APOE-ε4 status. While larger WMH and underweight predicted (p < 0.05) faster decreases in aHCV and cognition in APOE-ε4 non-carriers, lower plasma Aβ42/Aβ40 predicted (p < 0.031) faster decreases in aHCV and cognition in APOE-ε4 carriers. Higher Aβ PET also predicted faster rates of aHCV (p = 0.010) in APOE-ε4 carriers only, but was related to faster rates of cognitive decline (p < 0.022) regardless of APOE-ε4 status. These findings may provide novel insights into understanding different mechanisms underlie neurodegeneration and cognitive decline in non-demented elderly adults with and without APOE-ε4 allele, which may help the design of anti-Alzheimer's clinical trials.
Collapse
|
26
|
Pan FF, Huang Q, Wang Y, Wang YF, Guan YH, Xie F, Guo QH. Non-linear Character of Plasma Amyloid Beta Over the Course of Cognitive Decline in Alzheimer’s Continuum. Front Aging Neurosci 2022; 14:832700. [PMID: 35401142 PMCID: PMC8984285 DOI: 10.3389/fnagi.2022.832700] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/28/2022] [Indexed: 12/14/2022] Open
Abstract
Plasma amyloid-β (Aβ) was associated with brain Aβ deposition and Alzheimer’s disease (AD) development. However, changes of plasma Aβ over the course of cognitive decline in the Alzheimer’s continuum remained uncertain. We recruited 449 participants to this study, including normal controls (NC), subjective cognitive decline (SCD), mild cognitive impairment (MCI), AD, and non-AD dementia. All the participants underwent plasma Aβ42, Aβ40, and t-tau measurements with single-molecule array (Simoa) immunoassay and PET scan with 18F-florbetapir amyloid tracer. In the subgroup of Aβ-PET positive, plasma Aβ42 and Aβ42/Aβ40 ratio was significantly lower in AD than NC, SCD and MCI, yet SCD had significantly higher levels of plasma Aβ42 than both NC and MCI. In the diagnostic groups of MCI and dementia, participants with Aβ-PET positive had lower plasma Aβ42 and Aβ42/40 ratio than participants with Aβ-PET negative, and the increasing levels of plasma Aβ42 and Aβ42/40 ratio indicated lower risks of Aβ-PET positive. However, in the participants with SCD, plasma Aβ42 and Aβ40 were higher in the subgroup of Aβ-PET positive than Aβ-PET negative, and the increasing levels of plasma Aβ42 and Aβ40 indicated higher risks of Aβ-PET positive. No significant association was observed between plasma Aβ and Aβ-PET status in normal controls. These findings showed that, in the continuum of AD, plasma Aβ42 had a significantly increasing trend from NC to SCD before decreasing in MCI and AD. Furthermore, the predictive values of plasma Aβ for brain amyloid deposition were inconsistent over the course of cognitive decline.
Collapse
Affiliation(s)
- Feng-Feng Pan
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Qi Huang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Wang
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yi-Fan Wang
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yi-Hui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
- Fang Xie,
| | - Qi-Hao Guo
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- *Correspondence: Qi-Hao Guo,
| |
Collapse
|
27
|
Álvarez-Sánchez L, Peña-Bautista C, Baquero M, Cháfer-Pericás C. Novel Ultrasensitive Detection Technologies for the Identification of Early and Minimally Invasive Alzheimer's Disease Blood Biomarkers. J Alzheimers Dis 2022; 86:1337-1369. [PMID: 35213367 DOI: 10.3233/jad-215093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Single molecule array (SIMOA) and other ultrasensitive detection technologies have allowed the determination of blood-based biomarkers of Alzheimer's disease (AD) for diagnosis and monitoring, thereby opening up a promising field of research. OBJECTIVE To review the published bibliography on plasma biomarkers in AD using new ultrasensitive techniques. METHODS A systematic review of the PubMed database was carried out to identify reports on the use of blood-based ultrasensitive technology to identify biomarkers for AD. RESULTS Based on this search, 86 works were included and classified according to the biomarker determined. First, plasma amyloid-β showed satisfactory accuracy as an AD biomarker in patients with a high risk of developing dementia. Second, plasma t-Tau displayed good sensitivity in detecting different neurodegenerative diseases. Third, plasma p-Tau was highly specific for AD. Fourth, plasma NfL was highly sensitive for distinguishing between patients with neurodegenerative diseases and healthy controls. In general, the simultaneous determination of several biomarkers facilitated greater accuracy in diagnosing AD (Aβ42/Aβ40, p-Tau181/217). CONCLUSION The recent development of ultrasensitive technology allows the determination of blood-based biomarkers with high sensitivity, thus facilitating the early detection of AD through the analysis of easily obtained biological samples. In short, as a result of this knowledge, pre-symptomatic and early AD diagnosis may be possible, and the recruitment process for future clinical trials could be more precise. However, further studies are necessary to standardize levels of blood-based biomarkers in the general population and thus achieve reproducible results among different laboratories.
Collapse
Affiliation(s)
| | - Carmen Peña-Bautista
- Alzheimer Disease Research Group, Health Research Institute La Fe, Valencia, Spain
| | - Miguel Baquero
- Division of Neurology, University and Polytechnic Hospital La Fe, Valencia, Spain
| | | |
Collapse
|
28
|
Leuzy A, Mattsson‐Carlgren N, Palmqvist S, Janelidze S, Dage JL, Hansson O. Blood-based biomarkers for Alzheimer's disease. EMBO Mol Med 2022; 14:e14408. [PMID: 34859598 PMCID: PMC8749476 DOI: 10.15252/emmm.202114408] [Citation(s) in RCA: 179] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 11/02/2021] [Accepted: 11/05/2021] [Indexed: 12/01/2022] Open
Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD) represent a mounting public health challenge. As these diseases are difficult to diagnose clinically, biomarkers of underlying pathophysiology are playing an ever-increasing role in research, clinical trials, and in the clinical work-up of patients. Though cerebrospinal fluid (CSF) and positron emission tomography (PET)-based measures are available, their use is not widespread due to limitations, including high costs and perceived invasiveness. As a result of rapid advances in the development of ultra-sensitive assays, the levels of pathological brain- and AD-related proteins can now be measured in blood, with recent work showing promising results. Plasma P-tau appears to be the best candidate marker during symptomatic AD (i.e., prodromal AD and AD dementia) and preclinical AD when combined with Aβ42/Aβ40. Though not AD-specific, blood NfL appears promising for the detection of neurodegeneration and could potentially be used to detect the effects of disease-modifying therapies. This review provides an overview of the progress achieved thus far using AD blood-based biomarkers, highlighting key areas of application and unmet challenges.
Collapse
Affiliation(s)
- Antoine Leuzy
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityMalmöSweden
| | - Niklas Mattsson‐Carlgren
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityMalmöSweden
- Department of NeurologySkåne University HospitalLundSweden
- Wallenberg Centre for Molecular MedicineLund UniversityLundSweden
| | - Sebastian Palmqvist
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityMalmöSweden
- Memory ClinicSkåne University HospitalLundSweden
| | - Shorena Janelidze
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityMalmöSweden
| | - Jeffrey L Dage
- Stark Neuroscience Research InstituteIndiana University School of MedicineIndianapolisINUSA
| | - Oskar Hansson
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityMalmöSweden
- Memory ClinicSkåne University HospitalLundSweden
| |
Collapse
|
29
|
Teunissen CE, Verberk IMW, Thijssen EH, Vermunt L, Hansson O, Zetterberg H, van der Flier WM, Mielke MM, Del Campo M. Blood-based biomarkers for Alzheimer's disease: towards clinical implementation. Lancet Neurol 2021; 21:66-77. [PMID: 34838239 DOI: 10.1016/s1474-4422(21)00361-6] [Citation(s) in RCA: 492] [Impact Index Per Article: 123.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 12/13/2022]
Abstract
For many years, blood-based biomarkers for Alzheimer's disease seemed unattainable, but recent results have shown that they could become a reality. Convincing data generated with new high-sensitivity assays have emerged with remarkable consistency across different cohorts, but also independent of the precise analytical method used. Concentrations in blood of amyloid and phosphorylated tau proteins associate with the corresponding concentrations in CSF and with amyloid-PET or tau-PET scans. Moreover, other blood-based biomarkers of neurodegeneration, such as neurofilament light chain and glial fibrillary acidic protein, appear to provide information on disease progression and potential for monitoring treatment effects. Now the question emerges of when and how we can bring these biomarkers to clinical practice. This step would pave the way for blood-based biomarkers to support the diagnosis of, and development of treatments for, Alzheimer's disease and other dementias.
Collapse
Affiliation(s)
- Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands.
| | - Inge M W Verberk
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
| | - Elisabeth H Thijssen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
| | - Lisa Vermunt
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sölvegatan, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; UK Dementia Research Institute at UCL, London, UK; Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; Hong Kong Center for Neurodegenerative Diseases, Hong Kong Special Administrative Region, China
| | - Wiesje M van der Flier
- Alzheimer Center, Department of Neurology, and Department of Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
| | - Michelle M Mielke
- Department of Quantitative Health Sciences, and Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Marta Del Campo
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands; Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| |
Collapse
|
30
|
Pelgrim TA, Beran M, Twait EL, Geerlings MI, Vonk JM. Cross-sectional associations of tau protein biomarkers with semantic and episodic memory in older adults without dementia: A systematic review and meta-analysis. Ageing Res Rev 2021; 71:101449. [PMID: 34400308 DOI: 10.1016/j.arr.2021.101449] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/02/2021] [Accepted: 08/12/2021] [Indexed: 10/20/2022]
Abstract
Pathological tau is suggested to play a role in cognitive deterioration in the preclinical phase of Alzheimer's disease. We investigated cross-sectional associations of tau burden with episodic and semantic memory performance in older adults without dementia. A systematic search in MEDLINE (via PubMed), PsychINFO, and Embase resulted in 24 eligible studies for meta-analysis. Tau burden was assessed using CSF, PET, or histopathological measures. All studies evaluated associations of tau with episodic memory: weighted effect sizes were -0.46 (95 % CI [-0.73; -0.20], p < .001) for episodic composite scores, -0.19 ([-0.36; -0.03], p = .024) for delayed word list recall, and -0.05 ([-0.14; 0.04], p = .257) for logical memory. Fourteen studies evaluated associations of tau with semantic memory: weighted effect sizes were -0.28 ([-0.52; -0.04], p = .023) for semantic composite scores, -0.06 ([-0.16; 0.03], p = .194) for semantic fluency, and 0.06 ([-0.06; 0.18], p = .319) for picture naming. Our findings indicate that tau burden related to both episodic and semantic memory impairment in older individuals without a diagnosis of mild cognitive impairment or manifest dementia, with episodic composite scores showing the strongest association with tau burden. Future potential lies in developing more sensitive scores to detect this subtle cognitive impairment, which could contribute to early identification of individuals in the preclinical phase of Alzheimer's disease, thereby improving early diagnosis and timely intervention.
Collapse
|
31
|
Marcucci V, Kleiman J. Biomarkers and Their Implications in Alzheimer’s Disease: A Literature Review. EXPLORATORY RESEARCH AND HYPOTHESIS IN MEDICINE 2021; 000:000-000. [DOI: 10.14218/erhm.2021.00016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
32
|
Ding X, Zhang S, Jiang L, Wang L, Li T, Lei P. Ultrasensitive assays for detection of plasma tau and phosphorylated tau 181 in Alzheimer's disease: a systematic review and meta-analysis. Transl Neurodegener 2021; 10:10. [PMID: 33712071 PMCID: PMC7953695 DOI: 10.1186/s40035-021-00234-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/24/2021] [Indexed: 02/08/2023] Open
Abstract
A lack of convenient and reliable biomarkers for diagnosis and prognosis is a common challenge for neurodegenerative diseases such as Alzheimer's disease (AD). Recent advancement in ultrasensitive protein assays has allowed the quantification of tau and phosphorylated tau proteins in peripheral plasma. Here we identified 66 eligible studies reporting quantification of plasma tau and phosphorylated tau 181 (ptau181) using four ultrasensitive methods. Meta-analysis of these studies confirmed that the AD patients had significantly higher plasma tau and ptau181 levels compared with controls, and that the plasma tau and ptau181 could predict AD with high-accuracy area under curve of the Receiver Operating Characteristic. Therefore, plasma tau and plasma ptau181 can be considered as biomarkers for AD diagnosis.
Collapse
Affiliation(s)
- Xulong Ding
- Department of Neurology and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shuting Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lijun Jiang
- Mental Health Center and West China Brain Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lu Wang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tao Li
- Mental Health Center and West China Brain Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
33
|
Verberk IMW, Laarhuis MB, van den Bosch KA, Ebenau JL, van Leeuwenstijn M, Prins ND, Scheltens P, Teunissen CE, van der Flier WM. Serum markers glial fibrillary acidic protein and neurofilament light for prognosis and monitoring in cognitively normal older people: a prospective memory clinic-based cohort study. LANCET HEALTHY LONGEVITY 2021; 2:e87-e95. [DOI: 10.1016/s2666-7568(20)30061-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 01/11/2023]
|
34
|
Stockmann J, Verberk IMW, Timmesfeld N, Denz R, Budde B, Lange-Leifhelm J, Scheltens P, van der Flier WM, Nabers A, Teunissen CE, Gerwert K. Amyloid-β misfolding as a plasma biomarker indicates risk for future clinical Alzheimer's disease in individuals with subjective cognitive decline. Alzheimers Res Ther 2020; 12:169. [PMID: 33357241 PMCID: PMC7761044 DOI: 10.1186/s13195-020-00738-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/02/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND We evaluated Aβ misfolding in combination with Aβ42/40 ratio as a prognostic tool for future clinical progression to mild cognitive impairment (MCI) or dementia due to Alzheimer's disease (AD) in individuals with subjective cognitive decline (SCD). METHODS Baseline plasma samples (n = 203) from SCD subjects in the SCIENCe project and Amsterdam Dementia Cohort (age 61 ± 9 years; 57% male, mean follow-up time 2.7 years) were analyzed using immuno-infrared-sensor technology. Within 6 years of follow-up, 22 (11%) individuals progressed to MCI or dementia due to AD. Sensor readout values > 1646 cm- 1 reflected normal Aβ folding; readouts at ≤ 1646 cm- 1 reflected low and at < 1644 cm- 1 high misfolding. We used Cox proportional hazard models to quantify Aβ misfolding as a prognostic biomarker for progression to MCI and dementia due to AD. The accuracy of the predicted development of MCI/AD was determined by time-dependent receiver operating characteristic (t-ROC) curve analyses that take individual follow-up and conversion times into account. Statistical models were adjusted for age, sex, and APOEε4 status. Additionally, plasma Aβ42/40 data measured by SIMOA were statistically analyzed and compared. RESULTS All 22 patients who converted to MCI or AD-dementia within 6 years exhibited Aβ misfolding at baseline. Cox analyses revealed a hazard ratio (HR) of 19 (95% confidence interval [CI] 2.2-157.8) for future conversion of SCD subjects with high misfolding and of 11 (95% CI 1.0-110.1) for those with low misfolding. T-ROC curve analyses yielded an area under the curve (AUC) of 0.94 (95% CI 0.86-1.00; 6-year follow-up) for Aβ misfolding in an age, sex, and APOEε4 model. A similar model with plasma Aβ42/40 ratio yielded an AUC of 0.92 (95% CI, 0.82-1.00). The AUC increased to 0.99 (95% CI, 0.99-1.00) after inclusion of both Aβ misfolding and the Aβ42/40 ratio. CONCLUSIONS A panel of structure- and concentration-based plasma amyloid biomarkers may predict conversion to clinical MCI and dementia due to AD in cognitively unimpaired subjects. These plasma biomarkers provide a noninvasive and cost-effective alternative for screening early AD pathological changes. Follow-up studies and external validation in larger cohorts are in progress for further validation of our findings.
Collapse
Affiliation(s)
- Julia Stockmann
- Competence Center for Biospectroscopy, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr University Bochum, Faculty of Biology and Biotechnology, Bochum, Germany
| | - Inge M W Verberk
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Nina Timmesfeld
- Ruhr University Bochum, Department of Medical Informatics, Biometry and Epidemiology, Bochum, Germany
| | - Robin Denz
- Ruhr University Bochum, Department of Medical Informatics, Biometry and Epidemiology, Bochum, Germany
| | - Brian Budde
- Competence Center for Biospectroscopy, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr University Bochum, Faculty of Biology and Biotechnology, Bochum, Germany
| | - Julia Lange-Leifhelm
- Competence Center for Biospectroscopy, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr University Bochum, Faculty of Biology and Biotechnology, Bochum, Germany
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Andreas Nabers
- Competence Center for Biospectroscopy, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr University Bochum, Faculty of Biology and Biotechnology, Bochum, Germany
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Klaus Gerwert
- Competence Center for Biospectroscopy, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany.
- Department of Biophysics, Ruhr University Bochum, Faculty of Biology and Biotechnology, Bochum, Germany.
| |
Collapse
|
35
|
Giudici KV, de Souto Barreto P, Guyonnet S, Li Y, Bateman RJ, Vellas B. Assessment of Plasma Amyloid-β42/40 and Cognitive Decline Among Community-Dwelling Older Adults. JAMA Netw Open 2020; 3:e2028634. [PMID: 33331917 PMCID: PMC7747018 DOI: 10.1001/jamanetworkopen.2020.28634] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
IMPORTANCE Plasma measurement of amyloid-β (Aβ) peptides has been associated with cognitive function, but evidence of its ability to identify cognitive decline is still scarce. OBJECTIVE To investigate the associations between plasma Aβ42/40 and cognitive decline over time among community-dwelling older adults with subjective memory concerns. DESIGN, SETTING, AND PARTICIPANTS This multicenter cohort study used data from volunteers in the 5-year study Multidomain Alzheimer Preventive Trial (MAPT). Participants were aged 70 years or older and observed for a median (interquartile range) of 3.9 (2.0-4.0) years. Recruitment of participants started in May 2008 and ended in February 2011. Follow-up ended in April 2016. Data analysis was conducted from April to October 2020. EXPOSURE Plasma Aβ42 and Aβ40 were measured at 12 months for 448 participants (92.8%) and at 24 months for the rest. The moment of Aβ assessment was defined as the baseline for this study. MAIN OUTCOMES AND MEASURES Cognitive function was assessed at 12, 24, 36, 48, and 60 months by a composite cognitive score based on 4 tests; Mini Mental State Examination (MMSE); Clinical Dementia Rating, sum of boxes; and Alzheimer Disease Cooperative Study-Activities of Daily Living. Mixed-effect linear regressions were performed. RESULTS A total of 483 participants (median [IQR] age, 76.0 [73.0-80.0]; 286 [59.2%] women) were analyzed. Of them, 161 (33.3%) were classified as low plasma Aβ42/40 (≤0.107). After adjusting for age, sex, education, body mass index, Geriatric Depression Scale score, apolipoprotein E ε4 genotype, and MAPT intervention groups, low plasma Aβ42/40 was associated with more pronounced decline in composite cognitive score (adjusted between-group mean difference: -0.20, 95% CI, -0.34 to -0.07; P = .004) and decline in MMSE score (adjusted between-group mean difference: -0.59; 95% CI, -1.07 to -0.11; P = .02) during the follow-up period compared with the group with an Aβ42/40 ratio greater than 0.107. CONCLUSIONS AND RELEVANCE In this study, low plasma Aβ42/40 was associated with more pronounced decline in cognitive function (measured by multiple outcomes) over time. Findings suggest that plasma Aβ42/40 may be used to identify people at risk of cognitive decline, being an alternative to more complex and expensive measures, such as positron emission tomography imaging or cerebrospinal fluid measurement.
Collapse
Affiliation(s)
| | - Philipe de Souto Barreto
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital, Toulouse, France
- UPS/Inserm UMR1027, University of Toulouse III, Toulouse, France
| | - Sophie Guyonnet
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital, Toulouse, France
- UPS/Inserm UMR1027, University of Toulouse III, Toulouse, France
| | - Yan Li
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, Missouri
- Division of Biostatistics, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Randall John Bateman
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Bruno Vellas
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital, Toulouse, France
- UPS/Inserm UMR1027, University of Toulouse III, Toulouse, France
| |
Collapse
|
36
|
Vonk JMJ, Twait EL, Scholten RJPM, Geerlings MI. Cross-sectional associations of amyloid burden with semantic cognition in older adults without dementia: A systematic review and meta-analysis. Mech Ageing Dev 2020; 192:111386. [PMID: 33091462 PMCID: PMC7952036 DOI: 10.1016/j.mad.2020.111386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/13/2020] [Accepted: 10/14/2020] [Indexed: 12/31/2022]
Abstract
Previous research suggests the presence of subtle semantic decline in early stages of Alzheimer's disease. This study investigated associations between amyloid burden, a biomarker for Alzheimer's disease, and tasks of semantic impairment in older individuals without dementia. A systematic search in MEDLINE, PsycINFO, and Embase yielded 3691 peer-reviewed articles excluding duplicates. After screening, 41 studies with overall 7495 participants were included in the meta-analysis and quality assessment. The overall weighted effect size of the association between larger amyloid burden and larger semantic impairment was 0.10 (95% CI [-0.03; 0.22], p = 0.128) for picture naming, 0.19 (95% CI [0.11; 0.27], p < 0.001) for semantic fluency, 0.15 (95% CI [-0.15; 0.45], p = 0.326) for vocabulary, and 0.10 (95% CI [-0.14; 0.35], p = 0.405; 2 studies) for WAIS Information. Risk of bias was highest regarding comparability, as effect sizes were often not calculated on covariate-adjusted statistics. The relevance of the indicated amyloid-related decline in semantic fluency for research and clinical applications is likely negligible due to the effect's small magnitude. Future research should develop more sensitive metrics of semantic fluency to optimize its use for early detection of Alzheimer's disease-related cognitive impairment.
Collapse
Affiliation(s)
- Jet M J Vonk
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| | - Emma L Twait
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Rob J P M Scholten
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Mirjam I Geerlings
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| |
Collapse
|