1
|
Hellwig S, Frings L, Heibel M, Schroeter N, Blazhenets G, Domschke K, Brumberg J, Meyer PT. [ 18F]Fluorodeoxyglucose position emission tomography for differential diagnosis of depressive cognitive impairment: incremental value compared with clinical diagnosis. Br J Psychiatry 2025:1-8. [PMID: 40337859 DOI: 10.1192/bjp.2025.67] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
BACKGROUND Assessment of regional glucose metabolism by [18F]fluorodeoxyglucose position emission tomography ([18F]FDG PET) serves as a biomarker for differential diagnosis of dementia. Conversely, depressive cognitive impairment shows no abnormalities on cerebral [18F]FDG PET. AIMS This study validates the diagnostic value of [18F]FDG PET in addition to clinical diagnosis in a real-life gerontopsychiatric clinical population. METHOD Ninety-eight consecutive patients with depression and cognitive impairment were included. Baseline clinical diagnoses were independently established before and after disclosure of [18F]FDG PET, and dichotomised into neurodegenerative or non-neurodegenerative diseases (level 1). Subsequently, neurodegenerative cases were allocated to diagnostic subgroups (Alzheimer's disease, Lewy body diseases, frontotemporal lobar degeneration, neurodegenerative other; level 2). An interdisciplinary, biomarker-supported consensus diagnosis after a median follow-up of 6.6 month after [18F]FDG PET served as reference. Changes of clinical diagnoses and diagnostic accuracy were assessed. RESULTS After disclosure of [18F]FDG PET, level-1 clinical diagnoses changed in 23% (95% CI 16-33%) of cases, improving the diagnostic accuracy from 72% (95% CI 62-81%) to 92% (95% CI 84-96%) (P < 0.001). [18F]FDG PET was of particular value for exclusion of neurodegenerative disease. Concerning level-2 decisions, the clinical diagnoses changed in 30% (95% CI 21-40%) of cases, increasing its accuracy from 64% (95% CI 54-74%) to 85% (95% CI 76-91%) (P < 0.001). A major fraction of incorrect level-2 diagnoses comprised Alzheimer's disease misdiagnosed as Lewy body diseases. CONCLUSIONS [18F]FDG PET provides a significant incremental diagnostic value beyond the clinical diagnosis in depressive cognitive impairment. Thus, [18F]FDG PET should be considered in the diagnostic work-up of patients with mental disorders and cognitive impairment.
Collapse
Affiliation(s)
- Sabine Hellwig
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Lars Frings
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Meret Heibel
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Nils Schroeter
- Department of Neurology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Ganna Blazhenets
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Katharina Domschke
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- German Center for Mental Health (DZPG), Partner Site Berlin, Berlin, Germany
| | - Joachim Brumberg
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Philipp T Meyer
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
2
|
Kurz C, Carli L, Gürsel SÜ, Schrurs I, Jethwa A, Carboni M, Bittner T, Hortsch S, Keeser D, Brendel M, Burow L, Haeckert J, Koriath CAM, Tatò M, Utecht J, Papazov B, Morenas-Rodriguez E, Pogarell O, Palleis C, Weidinger E, Stoecklein S, Levin J, Höglinger G, Rauchmann BS, Perneczky R. Plasma biomarkers of amyloid, tau & neuroinflammation in Alzheimer's disease and corticobasal syndrome. Eur Arch Psychiatry Clin Neurosci 2025:10.1007/s00406-025-02013-z. [PMID: 40314736 DOI: 10.1007/s00406-025-02013-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 04/11/2025] [Indexed: 05/03/2025]
Abstract
Blood-based biomarkers (BBBMs) could significantly facilitate the diagnosis of Alzheimer's disease (AD) and non-AD dementia by providing less invasive alternatives to cerebrospinal fluid (CSF) and positron emission tomography (PET) imaging. This study investigated how well the BBBMs-amyloid-β (Aβ) 1-42/1-40 ratio, phosphorylated tau181 (pTau181), apolipoprotein E4 (ApoE4), glial fibrillary acidic protein (GFAP), and neurofilament light chain (NfL)-reflect thorough clinical work-up validated by PET and CSF biomarkers in participants with AD (n = 27), Aβ-negative CBS (n = 26), and age-matched healthy controls (HC) (n = 17). Factor and correlation explored biomarker associations. Bayesian regression, backward selection regression, and ROC curve analysis were applied to identify optimal biomarker combinations and diagnostic cut-offs. In AD cases, pTau181 and ApoE4 levels were elevated, and the Aβ1-42/1-40 ratio was reduced. ROC analysis showed high accuracy for pTau181, ApoE4 and Aβ1-42/1-40 in discriminating AD from HC, with a combination significantly improving performance. However, limited fold change, and high variability reduced the diagnostic applicability of Aβ1-42/1-40 ratio. Elevated NfL levels were the most reliable biomarker for CBS-Aβ(-) cases. GFAP showed limited discriminatory power due to overlapping levels, suggesting that it may not serve as a disease-specific biomarker but may be indicative of general neurodegeneration. This study highlights the diagnostic utility of pTau181, ApoE4 and the Aβ1-42/1-40 ratio for AD and NfL in the CBS-Aβ(-) cases and emphasizes the added value of combined biomarker models for group differentiation. Prospective studies will help validate these findings and refine clinical thresholds.
Collapse
Affiliation(s)
- Carolin Kurz
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstr. 7, 80336, Munich, Germany.
| | - Laura Carli
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstr. 7, 80336, Munich, Germany
| | - Selim Üstün Gürsel
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstr. 7, 80336, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377, Munich, Germany
| | - Isabelle Schrurs
- Roche Diagnostics International Ltd, 6343, Rotkreuz, Switzerland
| | | | | | | | | | - Daniel Keeser
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstr. 7, 80336, Munich, Germany
| | - Matthias Brendel
- Munich Cluster for Systems Neurology (SyNergy), 80336, Munich, Germany
- Department of Nuclear Medicine, LMU Hospital Munich, LMU Munich, 81377, Munich, Germany
| | - Lena Burow
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstr. 7, 80336, Munich, Germany
| | - Jan Haeckert
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstr. 7, 80336, Munich, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, 86156, Augsburg, Germany
| | - Carolin A M Koriath
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstr. 7, 80336, Munich, Germany
| | - Maia Tatò
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstr. 7, 80336, Munich, Germany
| | - Julia Utecht
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstr. 7, 80336, Munich, Germany
| | - Boris Papazov
- Clinic for Psychiatry, Psychotherapy and Psychosomatics at the University of Augsburg, Augsburg, Germany
| | - Estrella Morenas-Rodriguez
- German Center for Neurodegenerative Diseases (DZNE), 81377, Munich, Germany
- Institut de Recerca Hospital Sant Pau, 08041, Barcelona, Spain
| | - Oliver Pogarell
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstr. 7, 80336, Munich, Germany
| | - Carla Palleis
- German Center for Neurodegenerative Diseases (DZNE), 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 80336, Munich, Germany
- Department of Neurology, LMU Hospital Munich, LMU Munich, 81377, Munich, Germany
| | - Endy Weidinger
- Department of Neurology, LMU Hospital Munich, LMU Munich, 81377, Munich, Germany
| | - Sophia Stoecklein
- Department of Radiology, LMU Hospital Munich, LMU Munich, 81377, Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 80336, Munich, Germany
- Department of Neurology, LMU Hospital Munich, LMU Munich, 81377, Munich, Germany
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases (DZNE), 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 80336, Munich, Germany
- Department of Neurology, LMU Hospital Munich, LMU Munich, 81377, Munich, Germany
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstr. 7, 80336, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377, Munich, Germany
- Department of Radiology, LMU Hospital Munich, LMU Munich, 81377, Munich, Germany
- Department of Neuroradiology, LMU Hospital Munich, LMU Munich, 81377, Munich, Germany
| | - Robert Perneczky
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstr. 7, 80336, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 80336, Munich, Germany
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, W6 8RP, UK
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, S10 2HQ, UK
| |
Collapse
|
3
|
Thal DR, Poesen K, Vandenberghe R, De Meyer S. Alzheimer's disease neuropathology and its estimation with fluid and imaging biomarkers. Mol Neurodegener 2025; 20:33. [PMID: 40087672 PMCID: PMC11907863 DOI: 10.1186/s13024-025-00819-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
Alzheimer's disease (AD) is neuropathologically characterized by the extracellular deposition of the amyloid-β peptide (Aβ) and the intraneuronal accumulation of abnormal phosphorylated tau (τ)-protein (p-τ). Most frequently, these hallmark lesions are accompanied by other co-pathologies in the brain that may contribute to cognitive impairment, such as vascular lesions, intraneuronal accumulation of phosphorylated transactive-response DNA-binding protein 43 (TDP-43), and/or α-synuclein (αSyn) aggregates. To estimate the extent of these AD and co-pathologies in patients, several biomarkers have been developed. Specific tracers target and visualize Aβ plaques, p-τ and αSyn pathology or inflammation by positron emission tomography. In addition to these imaging biomarkers, cerebrospinal fluid, and blood-based biomarker assays reflecting AD-specific or non-specific processes are either already in clinical use or in development. In this review, we will introduce the pathological lesions of the AD brain, the related biomarkers, and discuss to what extent the respective biomarkers estimate the pathology determined at post-mortem histopathological analysis. It became evident that initial stages of Aβ plaque and p-τ pathology are not detected with the currently available biomarkers. Interestingly, p-τ pathology precedes Aβ deposition, especially in the beginning of the disease when biomarkers are unable to detect it. Later, Aβ takes the lead and accelerates p-τ pathology, fitting well with the known evolution of biomarker measures over time. Some co-pathologies still lack clinically established biomarkers today, such as TDP-43 pathology or cortical microinfarcts. In summary, specific biomarkers for AD-related pathologies allow accurate clinical diagnosis of AD based on pathobiological parameters. Although current biomarkers are excellent measures for the respective pathologies, they fail to detect initial stages of the disease for which post-mortem analysis of the brain is still required. Accordingly, neuropathological studies remain essential to understand disease development especially in early stages. Moreover, there is an urgent need for biomarkers reflecting co-pathologies, such as limbic predominant, age-related TDP-43 encephalopathy-related pathology, which is known to modify the disease by interacting with p-τ. Novel biomarker approaches such as extracellular vesicle-based assays and cryptic RNA/peptides may help to better detect these co-pathologies in the future.
Collapse
Affiliation(s)
- Dietmar Rudolf Thal
- Department of Imaging and Pathology, Laboratory for Neuropathology, Leuven Brain Institute, KU Leuven, Herestraat 49, Leuven, 3000, Belgium.
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium.
| | - Koen Poesen
- Department of Neurosciences, Laboratory for Molecular Neurobiomarker Research, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Department of Neurosciences, Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Steffi De Meyer
- Department of Neurosciences, Laboratory for Molecular Neurobiomarker Research, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Neurosciences, Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
4
|
Leuzy A, Bollack A, Pellegrino D, Teunissen CE, La Joie R, Rabinovici GD, Franzmeier N, Johnson K, Barkhof F, Shaw LM, Arkhipenko A, Schindler SE, Honig LS, Moscoso Rial A, Schöll M, Zetterberg H, Blennow K, Hansson O, Farrar G. Considerations in the clinical use of amyloid PET and CSF biomarkers for Alzheimer's disease. Alzheimers Dement 2025; 21:e14528. [PMID: 40042435 PMCID: PMC11881640 DOI: 10.1002/alz.14528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/21/2024] [Accepted: 12/06/2024] [Indexed: 03/09/2025]
Abstract
Amyloid-β (Aβ) positron emission tomography (PET) imaging and cerebrospinal fluid (CSF) biomarkers are now established tools in the diagnostic workup of patients with Alzheimer's disease (AD), and their use is anticipated to increase with the introduction of new disease-modifying therapies. Although these biomarkers are comparable alternatives in research settings to determine Aβ status, biomarker testing in clinical practice requires careful consideration of the strengths and limitations of each modality, as well as the specific clinical context, to identify which test is best suited for each patient. This article provides a comprehensive review of the pathologic processes reflected by Aβ-PET and CSF biomarkers, their performance, and their current and future applications and contexts of use. The primary aim is to assist clinicians in making better-informed decisions about the suitability of each biomarker in different clinical situations, thereby reducing the risk of misdiagnosis or incorrect interpretation of biomarker results. HIGHLIGHTS: Recent advances have positioned Aβ PET and CSF biomarkers as pivotal in AD diagnosis. It is crucial to understand the differences in the clinical use of these biomarkers. A team of experts reviewed the state of Aβ PET and CSF markers in clinical settings. Differential features in the clinical application of these biomarkers were reviewed. We discussed the role of Aβ PET and CSF in the context of novel plasma biomarkers.
Collapse
Grants
- AF-930351 Neurodegenerative Disease Research
- 101053962 National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
- R01 AG066107 NIA NIH HHS
- FO2022-0270 Bluefield Project, Olav Thon Foundation, Erling-Persson Family Foundation
- 101112145 European Union's Horizon Europe
- Alzheimer Netherlands
- ZEN-21-848495 Alzheimer's Association 2021 Zenith Award
- 2022-0231 Knut and Alice Wallenberg foundation
- KAW 2023.0371 Knut and Alice Wallenberg Foundation
- U19 ADNI4 Harvard Aging Brain Study
- R01 AG081394 NIA NIH HHS
- ADRC P30-AG-072979 Harvard Aging Brain Study
- 2022-1259 Regionalt Forskningsstöd
- Shanendoah Foundation
- 2020-O000028 Konung Gustaf V:s och Drottning Victorias Frimurarestiftelse, Skåne University Hospital Foundation
- The Selfridges Group Foundation
- R56 AG057195 NIA NIH HHS
- U01 NS100600 NINDS NIH HHS
- ALZ2022-0006 Hjärnfonden, Sweden
- U01 AG057195 NIA NIH HHS
- Dutch National Dementia Strategy
- ZEN24-1069572 Alzheimer's Association
- R01AG072474 Harvard Aging Brain Study
- 860197 Marie Curie International Training Network
- AF-939721 Neurodegenerative Disease Research
- R01 AG070941 NIA NIH HHS
- P01 AG036694 NIA NIH HHS
- JPND2021-00694 Neurodegenerative Disease Research
- ADSF-21-831376-C AD Strategic Fund, and Alzheimer's Association
- AF-994900 Swedish Alzheimer Foundation
- NIH
- ALFGBG-813971 County Councils, the ALF-agreement
- FO2021-0293 Swedish Brain Foundation
- U19AG063893 NINDS NIH HHS
- 2022-01018 National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
- 201809-2016862 National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
- 831434 Innovative Medicines Initiatives 3TR
- 101132933 European Union's Horizon Europe
- European Union Joint Programme
- Cure Alzheimer's fund, Rönström Family Foundation
- ID 390857198 Munich Cluster for Systems Neurology
- U01-AG057195 NIA NIH HHS
- Deutsche Forschungsgemeinschaft
- 2021-06545 Swedish Research Council
- Sahlgrenska Academy at the University of Gothenburg
- U19 AG024904 NIA NIH HHS
- GE Healthcare
- JPND2019-466-236 European Union Joint Program for Neurodegenerative Disorders
- P30 AG062422 NIA NIH HHS
- ADG-101096455 European Research Council
- 2022-00732 Neurodegenerative Disease Research
- 860197 Marie Skłodowska-Curie
- P01 AG019724 NIA NIH HHS
- U01NS100600 NINDS NIH HHS
- AF-980907 Strategic Research Area MultiPark (Multidisciplinary Research in Parkinson's disease) at Lund University, Swedish Alzheimer Foundation
- P30 AG066462 NIA NIH HHS
- 2022-00775 GHR Foundation, Swedish Research Council
- R44 AG071388 NIA NIH HHS
- FO2017-0243 Hjärnfonden, Sweden
- AF-968270 Neurodegenerative Disease Research
- KAW2014.0363 Knut and Alice Wallenberg Foundation
- SG-23-1061717 Alzheimer's Association
- 2021-02678 Swedish Research Council
- R01 AG059013 NIA NIH HHS
- R35 AG072362 NIA NIH HHS
- VGFOUREG-995510 Västra Götaland Region R&D
- American College of Radiology
- R01 AG081394-01 European Union's Horizon Europe
- R21 AG070768 NIA NIH HHS
- U19 AG063893 NIA NIH HHS
- 2022-Projekt0080 Swedish Federal Government under the ALF agreement
- ALFGBG-965326 County Councils, the ALF-agreement
- Alzheimer Drug Discovery Foundation
- Rainwater Charitable Foundation
- Research of the European Commission
- R01AG083740 National Institute of Aging
- ADSF-21-831381-C AD Strategic Fund, and Alzheimer's Association
- SG-23-1038904 Alzheimer's Association 2022-2025
- RS-2023-00263612 National Research Foundation of Korea
- P30-AG062422 NIA NIH HHS
- R21AG070768 Harvard Aging Brain Study
- 2017-02869 Swedish Research Council
- 101034344 Joint Undertaking
- ALFGBG-715986 Swedish state under the agreement between the Swedish government and the County Councils, ALF-agreement
- ERAPERMED2021-184 ERA PerMed
- U19AG024904 Harvard Aging Brain Study
- R01 AG072474 NIA NIH HHS
- UKDRI-1003 Neurodegenerative Disease Research
- 10510032120003 Health Holland, the Dutch Research Council
- 2019-02397 National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
- EXC 2145 SyNergy Munich Cluster for Systems Neurology
- 1412/22 Parkinson foundation of Sweden
- R01 AG046396 NIA NIH HHS
- ALFGBG-71320 National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
- P01-AG019724 NIA NIH HHS
- ALFGBG-965240 Swedish state under the agreement between the Swedish government and the County Councils, ALF-agreement
- Deutsche Parkinson Gesellschaft
- ADSF-21-831377-C AD Strategic Fund, and Alzheimer's Association
- National MS Society
- R01 AG083740 NIA NIH HHS
- 2017-00915 Neurodegenerative Disease Research
- 2023-06188 Swedish Research Council
- Alzheimer Association
- National MS Society
- Alzheimer Netherlands
- NIH
- NIA
- National Institute of Neurological Disorders and Stroke
- American College of Radiology
- Rainwater Charitable Foundation
- Deutsche Forschungsgemeinschaft
- NINDS
- Knut and Alice Wallenberg Foundation
- Swedish Research Council
- National Research Foundation of Korea
- Swedish Brain Foundation
- European Research Council
- Alzheimer's Association
- GE Healthcare
Collapse
Affiliation(s)
- Antoine Leuzy
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityLundSweden
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- The Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Department of NeuropsychiatrySahlgrenska University HospitalRegion Västra GötalandGothenburgSweden
| | - Ariane Bollack
- The Grove CentreWhite Lion Road BuckinghamshireGE HealthCareAmershamUK
- Department of Medical Physics and BioengineeringCentre for Medical Image Computing (CMIC)University College LondonLondonUK
| | | | - Charlotte E. Teunissen
- Neurochemistry LaboratoryDepartment of Laboratory MedicineAmsterdam NeuroscienceNeurodegenerationAmsterdam UMC Vrije UniversiteitAmsterdamThe Netherlands
| | - Renaud La Joie
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Gil D. Rabinovici
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Nicolai Franzmeier
- The Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Institute for Stroke and Dementia Research (ISD)University HospitalLMU MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Keith Johnson
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Center for Alzheimer Research and TreatmentBrigham and Women's HospitalBostonMassachusettsUSA
| | - Frederik Barkhof
- Department of Radiology and Nuclear MedicineVrije Universiteit AmsterdamAmsterdam University Medical CenterAmsterdamThe Netherlands
- Amsterdam NeuroscienceBrain imagingAmsterdamThe Netherlands
- UCL Queen Square Institute of Neurology and Center for Medical Image ComputingUniversity College LondonLondonUK
| | - Leslie M. Shaw
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Suzanne E. Schindler
- Department of NeurologyKnight Alzheimer's Disease Research CenterWashington University School of MedicineSt. LouisMissouriUSA
| | - Lawrence S. Honig
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Alexis Moscoso Rial
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- The Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Nuclear Medicine Department and Molecular Imaging GroupInstituto de Investigación Sanitaria de Santiago de CompostelaSantiago de CompostelaSpain
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- The Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Department of NeuropsychiatrySahlgrenska University HospitalRegion Västra GötalandGothenburgSweden
- Dementia Research CentreInstitute of NeurologyUniversity College LondonLondonUK
| | - Henrik Zetterberg
- The Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseQueen Square Institute of NeurologyUniversity College LondonLondonUK
- UK Dementia Research InstituteUniversity College LondonLondonUK
- Hong Kong Center for Neurodegenerative DiseasesScience ParkHong KongChina
- Wisconsin Alzheimer's Disease Research CenterSchool of Medicine and Public HealthUniversity of WisconsinUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Kaj Blennow
- The Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Paris Brain InstituteICMPitié‐Salpêtrière HospitalSorbonne UniversityParisFrance
- Neurodegenerative Disorder Research CenterDivision of Life Sciences and Medicineand Department of NeurologyInstitute on Aging and Brain DisordersUniversity of Science and Technology of China and First Affiliated Hospital of USTCHefeiChina
| | - Oskar Hansson
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityLundSweden
- Memory ClinicSkåne University HospitalMalmöSweden
| | - Gill Farrar
- The Grove CentreWhite Lion Road BuckinghamshireGE HealthCareAmershamUK
| |
Collapse
|
5
|
Dai C, Xu Y, Ke L, Zhu M, Deng R, Wang X, Zhou Y. Multiple-Signal Amplification Strategy to Fabricate an Ultrasensitive Electrochemiluminescence Magnetic Immunosensor for Detecting Biomarkers of Alzheimer's Disease via Iridium-Based Self-Enhancing Nanoemitters. ACS Sens 2025; 10:1083-1092. [PMID: 39835816 DOI: 10.1021/acssensors.4c02916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Alzheimer's disease (AD) is characterized by progressive memory loss and cognitive decline, significantly impairing the daily life of elderly individuals. The low abundance of blood-based biomarkers in AD necessitates higher analytical technique requirements. Herein, one novel iridium-based ECL self-enhanced nanoemitter (TPrA@Ir-SiO2) was unprecedentedly reported, and it was further used to construct an ultrasensitive ECL magnetic immunosensor by a multiple-signal amplification strategy to unequally sensitively and accurately detect the AD blood-based biomarker (P-tau181) in this work. The initial signal amplification was accomplished via incorporating a new efficient iridium-based luminophore named Ir(mdq)2(acac) and a corresponding coreactant into silica nanoparticles to successfully obtain TPrA@Ir-SiO2. In addition, the specific and high-affinity interactions between streptavidin and biotin were subsequently employed to further facilitate signal amplification. Based on the advantages of the luminophore itself and the high-affinity interactions between biotin and streptavidin, the corresponding ECL immunosensor proposed in this work exhibited remarkable sensitivity, covering a wide linear range from 0.1 pg/mL to 0.1 μg/mL, and achieved an ultralow limit of detection of 68.58 fg/mL (S/N = 3), and it also exhibited outstanding recovery (98-104%) and RSD (1.92-4.86%) in the detection of serum samples by the spiking method. These remarkable results undoubtedly demonstrate the potential of self-enhanced ECL nanoemitters combined with a synergistic signal amplification strategy bearing streptavidin-biotin in detecting AD blood-based biomarkers, providing accurate and reliable solutions for early diagnosis and monitoring of AD, which would open a new avenue to effectively reduce the burden on AD patients' families and society in the future.
Collapse
Affiliation(s)
- Chenji Dai
- School of Chemistry and Life Sciences, Jiangsu Key Laboratory for Environmental Functional Materials, Suzhou University of Science and Technology, Suzhou, Jiangsu 215009, China
| | - Yaoyao Xu
- School of Chemistry and Life Sciences, Jiangsu Key Laboratory for Environmental Functional Materials, Suzhou University of Science and Technology, Suzhou, Jiangsu 215009, China
| | - Libing Ke
- School of Chemistry and Life Sciences, Jiangsu Key Laboratory for Environmental Functional Materials, Suzhou University of Science and Technology, Suzhou, Jiangsu 215009, China
| | - Mengjiao Zhu
- School of Chemistry and Life Sciences, Jiangsu Key Laboratory for Environmental Functional Materials, Suzhou University of Science and Technology, Suzhou, Jiangsu 215009, China
| | - Rongxiu Deng
- School of Chemistry and Life Sciences, Jiangsu Key Laboratory for Environmental Functional Materials, Suzhou University of Science and Technology, Suzhou, Jiangsu 215009, China
| | - Xuedong Wang
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Yuyang Zhou
- School of Chemistry and Life Sciences, Jiangsu Key Laboratory for Environmental Functional Materials, Suzhou University of Science and Technology, Suzhou, Jiangsu 215009, China
| |
Collapse
|
6
|
Khalafi M, Dartora WJ, McIntire LBJ, Butler TA, Wartchow KM, Hojjati SH, Razlighi QR, Shirbandi K, Zhou L, Chen K, Xi K, Banerjee S, Foldi N, Pahlajani S, Glodzik L, Li Y, de Leon MJ, Chiang GC. Diagnostic accuracy of phosphorylated tau217 in detecting Alzheimer's disease pathology among cognitively impaired and unimpaired: A systematic review and meta-analysis. Alzheimers Dement 2025; 21:e14458. [PMID: 39711334 PMCID: PMC11848338 DOI: 10.1002/alz.14458] [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: 08/30/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024]
Abstract
Our review summarizes the diagnostic accuracy of plasma and cerebrospinal fluid (CSF) phosphorylated tau 217 (p-tau217) in detecting amyloid and tau pathology on positron emission tomography (PET). We systematically reviewed studies that reported the diagnostic accuracy of plasma and CSF p-tau217, searching MEDLINE/PubMed, Scopus, and Web of Science through August 2024. The accuracy of p-tau217 in predicting amyloid and tau pathology on PET was evaluated in 30 studies. Both plasma and CSF p-tau217 effectively detect amyloid and tau PET deposition. Plasma p-tau217 showed 82% sensitivity for detecting amyloid and 83% for tau, with 86% and 83% specificity, respectively. CSF p-tau217 had 79% sensitivity for amyloid and 91% for tau, with 91% and 84% specificity. p-tau217 effectively identifies Alzheimer's disease (AD) pathology. Plasma p-tau217 was comparable to CSF p-tau217 in detecting amyloid deposition on PET. Despite being less sensitive for detecting tau deposition on PET, plasma p-tau217 can be an efficient screening tool for underlying AD pathology. HIGHLIGHTS: Plasma phosphorylated tau 217 (p-tau217) serves as a viable biomarker alternative to cerebrospinal fluid p-tau217 due to the strong concordance between their results. Plasma p-tau217 accurately identifies amyloid and tau positron emission tomography (PET) positivity, exhibiting a low rate of false negatives and positives, thereby establishing it as a reliable diagnostic tool for Alzheimer's disease (AD). Plasma p-tau217 demonstrates slightly higher accuracy in predicting amyloid PET positivity compared to tau PET positivity. Plasma p-tau217 demonstrates higher predictive accuracy in detecting AD pathology among cognitively impaired individuals, compared to cognitively unimpaired individuals, suggesting its enhanced utility as a diagnostic biomarker in clinical settings.
Collapse
Affiliation(s)
- Mohammad Khalafi
- Department of RadiologyBrain Health Imaging InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - William J. Dartora
- Lipidomics and Biomarker Discovery LabDepartment of RadiologyBrain Health Imaging InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - Laura Beth J. McIntire
- Lipidomics and Biomarker Discovery LabDepartment of RadiologyBrain Health Imaging InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - Tracy A. Butler
- Department of RadiologyBrain Health Imaging InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - Krista M. Wartchow
- Lipidomics and Biomarker Discovery LabDepartment of RadiologyBrain Health Imaging InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - Seyed Hani Hojjati
- Department of RadiologyBrain Health Imaging InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - Qolamreza R. Razlighi
- Department of RadiologyBrain Health Imaging InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - Kiarash Shirbandi
- Biomedical Engineering DepartmentScience and Research BranchIslamic Azad UniversityTehranIran
| | - Liangdong Zhou
- Department of RadiologyBrain Health Imaging InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - Kewei Chen
- Banner Alzheimer's InstituteBanner HealthPhoenixArizonaUSA
- College of Health SolutionsArizona State UniversityPhoenixArizonaUSA
| | - Ke Xi
- Department of RadiologyBrain Health Imaging InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - Samprit Banerjee
- Department of Population Health SciencesWeill Cornell MedicineNew YorkNew YorkUSA
| | - Nancy Foldi
- Department of RadiologyBrain Health Imaging InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - Silky Pahlajani
- Department of RadiologyBrain Health Imaging InstituteWeill Cornell MedicineNew YorkNew YorkUSA
- Department of NeurologyWeill Cornell MedicineNew YorkNew YorkUSA
| | - Lidia Glodzik
- Department of RadiologyBrain Health Imaging InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - Yi Li
- Department of RadiologyBrain Health Imaging InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - Mony J. de Leon
- Department of RadiologyBrain Health Imaging InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - Gloria C. Chiang
- Department of RadiologyBrain Health Imaging InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| |
Collapse
|
7
|
Lohman T, Sible I, Engstrom AC, Kapoor A, Shenasa F, Head E, Sordo L, Alitin JPM, Gaubert A, Nguyen A, Rodgers KE, Bradford D, Nation DA. Beat-to-beat blood pressure variability, hippocampal atrophy, and memory impairment in older adults. GeroScience 2025; 47:993-1003. [PMID: 39098984 PMCID: PMC11872826 DOI: 10.1007/s11357-024-01303-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/23/2024] [Indexed: 08/06/2024] Open
Abstract
Visit-to-visit blood pressure variability (BPV) predicts age-related hippocampal atrophy, neurodegeneration, and memory decline in older adults. Beat-to-beat BPV may represent a more reliable and efficient tool for prospective risk assessment, but it is unknown whether beat-to-beat BPV is similarly associated with hippocampal neurodegeneration, or with plasma markers of neuroaxonal/neuroglial injury. Independently living older adults without a history of dementia, stroke, or other major neurological disorders were recruited from the community (N = 104; age = 69.5 ± 6.7 (range 55-89); 63% female). Participants underwent continuous blood pressure monitoring, brain MRI, venipuncture, and cognitive testing over two visits. Hippocampal volumes, plasma neurofilament light, and glial fibrillary acidic protein levels were assessed. Beat-to-beat BPV was quantified as systolic blood pressure average real variability during 7-min of supine continuous blood pressure monitoring. The cross-sectional relationship between beat-to-beat BPV and hippocampal volumes, cognitive domain measures, and plasma biomarkers was assessed using multiple linear regression with adjustment for demographic covariates, vascular risk factors, and average systolic blood pressure. Elevated beat-to-beat BPV was associated with decreased left hippocampal volume (P = .008), increased plasma concentration of glial fibrillary acidic protein (P = .006), and decreased memory composite score (P = .02), independent of age, sex, average systolic blood pressure, total intracranial volume, and vascular risk factor burden. In summary, beat-to-beat BPV is independently associated with decreased left hippocampal volume, increased neuroglial injury, and worse memory ability. Findings are consistent with prior studies examining visit-to-visit BPV and suggest beat-to-beat BPV may be a useful marker of hemodynamic brain injury in older adults.
Collapse
Affiliation(s)
- Trevor Lohman
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Isabel Sible
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Allison C Engstrom
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Arunima Kapoor
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Fatemah Shenasa
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | - Lorena Sordo
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | - John Paul M Alitin
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Aimee Gaubert
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Amy Nguyen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Kathleen E Rodgers
- Center for Innovations in Brain Science, Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - David Bradford
- Center for Innovations in Brain Science, Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Daniel A Nation
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
8
|
Spitz G, Hicks AJ, McDonald SJ, Dore V, Krishnadas N, O’Brien TJ, O’Brien WT, Vivash L, Law M, Ponsford JL, Rowe C, Shultz SR. Plasma biomarkers in chronic single moderate-severe traumatic brain injury. Brain 2024; 147:3690-3701. [PMID: 39315931 PMCID: PMC11531850 DOI: 10.1093/brain/awae255] [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: 08/13/2023] [Revised: 06/06/2024] [Accepted: 07/04/2024] [Indexed: 09/25/2024] Open
Abstract
Blood biomarkers are an emerging diagnostic and prognostic tool that reflect a range of neuropathological processes following traumatic brain injury (TBI). Their effectiveness in identifying long-term neuropathological processes after TBI is unclear. Studying biomarkers in the chronic phase is vital because elevated levels in TBI might result from distinct neuropathological mechanisms during acute and chronic phases. Here, we examine plasma biomarkers in the chronic period following TBI and their association with amyloid and tau PET, white matter microarchitecture, brain age and cognition. We recruited participants ≥40 years of age who had suffered a single moderate-severe TBI ≥10 years previously between January 2018 and March 2021. We measured plasma biomarkers using single molecule array technology [ubiquitin C-terminal hydrolase L1 (UCH-L1), neurofilament light (NfL), tau, glial fibrillary acidic protein (GFAP) and phosphorylated tau (P-tau181)]; PET tracers to measure amyloid-β (18F-NAV4694) and tau neurofibrillary tangles (18F-MK6240); MRI to assess white matter microstructure and brain age; and the Rey Auditory Verbal Learning Test to measure verbal-episodic memory. A total of 90 post-TBI participants (73% male; mean = 58.2 years) were recruited on average 22 years (range = 10-33 years) post-injury, and 32 non-TBI control participants (66% male; mean = 57.9 years) were recruited. Plasma UCH-L1 levels were 67% higher {exp(b) = 1.67, P = 0.018, adjusted P = 0.044, 95% confidence interval (CI) [10% to 155%], area under the curve = 0.616} and P-tau181 were 27% higher {exp(b) = 1.24, P = 0.011, adjusted P = 0.044, 95% CI [5% to 46%], area under the curve = 0.632} in TBI participants compared with controls. Amyloid and tau PET were not elevated in TBI participants. Higher concentrations of plasma P-tau181, UCH-L1, GFAP and NfL were significantly associated with worse white matter microstructure but not brain age in TBI participants. For TBI participants, poorer verbal-episodic memory was associated with higher concentration of P-tau181 {short delay: b = -2.17, SE = 1.06, P = 0.043, 95% CI [-4.28, -0.07]; long delay: bP-tau = -2.56, SE = 1.08, P = 0.020, 95% CI [-4.71, -0.41]}, tau {immediate memory: bTau = -6.22, SE = 2.47, P = 0.014, 95% CI [-11.14, -1.30]} and UCH-L1 {immediate memory: bUCH-L1 = -2.14, SE = 1.07, P = 0.048, 95% CI [-4.26, -0.01]}, but was not associated with functional outcome. Elevated plasma markers related to neuronal damage and accumulation of phosphorylated tau suggest the presence of ongoing neuropathology in the chronic phase following a single moderate-severe TBI. Plasma biomarkers were associated with measures of microstructural brain disruption on MRI and disordered cognition, further highlighting their utility as potential objective tools to monitor evolving neuropathology post-TBI.
Collapse
Affiliation(s)
- Gershon Spitz
- Monash-Epworth Rehabilitation Research Centre, School of Psychological Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3800, Australia
- Department of Neuroscience, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3004, Australia
| | - Amelia J Hicks
- Monash-Epworth Rehabilitation Research Centre, School of Psychological Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Stuart J McDonald
- Department of Neuroscience, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3004, Australia
- Department of Neurology, The Alfred, Melbourne, VIC 3004, Australia
| | - Vincent Dore
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC 3084, Australia
| | - Natasha Krishnadas
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC 3084, Australia
| | - Terence J O’Brien
- Department of Neuroscience, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3004, Australia
- Department of Neurology, The Alfred, Melbourne, VIC 3004, Australia
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3010, Australia
| | - William T O’Brien
- Department of Neuroscience, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3004, Australia
| | - Lucy Vivash
- Department of Neuroscience, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3004, Australia
- Department of Neurology, The Alfred, Melbourne, VIC 3004, Australia
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Meng Law
- Department of Neuroscience, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3004, Australia
- Department of Radiology, Alfred Health, Melbourne, VIC 3004, Australia
| | - Jennie L Ponsford
- Monash-Epworth Rehabilitation Research Centre, School of Psychological Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Christopher Rowe
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC 3084, Australia
| | - Sandy R Shultz
- Department of Neuroscience, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3004, Australia
- Department of Neurology, The Alfred, Melbourne, VIC 3004, Australia
- The Centre for Trauma and Mental Health Research, Health Sciences and Human Services, Vancouver Island University, Nanaimo, BC V9R 5S5, Canada
| |
Collapse
|
9
|
Kamatham PT, Shukla R, Khatri DK, Vora LK. Pathogenesis, diagnostics, and therapeutics for Alzheimer's disease: Breaking the memory barrier. Ageing Res Rev 2024; 101:102481. [PMID: 39236855 DOI: 10.1016/j.arr.2024.102481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/28/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and accounts for 60-70 % of all cases. It affects millions of people worldwide. AD poses a substantial economic burden on societies and healthcare systems. AD is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired daily functioning. As the prevalence of AD continues to increase, understanding its pathogenesis, improving diagnostic methods, and developing effective therapeutics have become paramount. This comprehensive review delves into the intricate mechanisms underlying AD, explores the current state of diagnostic techniques, and examines emerging therapeutic strategies. By revealing the complexities of AD, this review aims to contribute to the growing body of knowledge surrounding this devastating disease.
Collapse
Affiliation(s)
- Pushpa Tryphena Kamatham
- Molecular and Cellular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Rashi Shukla
- Molecular and Cellular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Dharmendra Kumar Khatri
- Department of Pharmacology, Nims Institute of Pharmacy, Nims University Rajasthan, Jaipur, India.
| | - Lalitkumar K Vora
- School of Pharmacy, Medical Biology Centre, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland BT9 7BL, UK.
| |
Collapse
|
10
|
Leuzy A, Heeman F, Bosch I, Lenér F, Dottori M, Quitz K, Moscoso A, Kern S, Zetterberg H, Blennow K, Schöll M. REAL AD-Validation of a realistic screening approach for early Alzheimer's disease. Alzheimers Dement 2024; 20:8172-8182. [PMID: 39311530 PMCID: PMC11567841 DOI: 10.1002/alz.14219] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 11/17/2024]
Abstract
Early diagnosis is crucial to treatment success. This is especially relevant for Alzheimer's disease (AD), with its protracted preclinical phase. Most health care systems do not have the resources to conduct large-scale AD screenings in middle-aged individuals in need of novel AD treatment options and early, accurate diagnosis. Recent developments in blood-based biomarkers and remote cognitive testing offer novel, cost-effective, and scalable methods to detect cognitive and biomarker changes that may indicate early AD. In research cohorts, promising results have been reported, but these modalities have not been validated in population-based settings. The validation of a realistic screening approach for early Alzheimer's disease (REAL AD) study aims to validate the diagnostic and prognostic performance of the combined use of blood-based biomarkers and remote cognitive testing as a screening approach for early AD employing an existing health care infrastructure (the Swedish Västra Götaland Region Primary Healthcare). REAL AD aims to provide a concrete, individualized diagnostic framework, which could significantly improve AD prognosis. HIGHLIGHTS: In Sweden, most Alzheimer's disease (AD) diagnoses are made in primary care, where access to AD biomarkers is almost non-existent. Most health care systems have limited resources for the screening of middle-aged adults for early evidence of AD pathology. Blood-based biomarkers and remote cognitive testing offer novel, cost-effective, and scalable methods for detecting cognitive and biomarker changes that may indicate early AD. The REAL AD study aims to validate the diagnostic and prognostic performance of blood-based biomarkers and remote cognitive testing as a screening approach for early AD in an existing primary health care infrastructure in the Västra Götaland Region in Sweden. Studies such as REAL AD will play a vital role in helping to move the field toward concrete implementation of biomarkers in AD diagnostic workup at all care levels, eventually providing more comprehensive treatments options for the large and growing AD population, and for those at risk.
Collapse
Affiliation(s)
- Antoine Leuzy
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- Department of Psychiatry and NeurochemistryUniversity of GothenburgMölndalSweden
- Department of NeuropsychiatryRegion Västra GötalandSahlgrenska University HospitalGötalandSweden
| | - Fiona Heeman
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- Department of Psychiatry and NeurochemistryUniversity of GothenburgMölndalSweden
| | - Iris Bosch
- Department of Psychiatry and NeurochemistryUniversity of GothenburgMölndalSweden
- Department of NeuropsychiatryRegion Västra GötalandSahlgrenska University HospitalGötalandSweden
| | - Frida Lenér
- Centre for REDI FyrbodalPrimary Health Care, Region VästraGötalandSweden
- Department of Public Health and Community MedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Maria Dottori
- Region Västra Götaland, Research, Education, Development & Innovation (REDI)Primary Health CareGothenburgSweden
| | - Kajsa Quitz
- Department of Public Health and Community MedicineUniversity of GothenburgGothenburgSweden
| | - Alexis Moscoso
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- Department of Psychiatry and NeurochemistryUniversity of GothenburgMölndalSweden
| | - Silke Kern
- Department of Psychiatry and NeurochemistryUniversity of GothenburgMölndalSweden
- Department of NeuropsychiatryRegion Västra GötalandSahlgrenska University HospitalGötalandSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryUniversity of GothenburgMölndalSweden
- Department of Public Health and Community MedicineUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalGothenburgSweden
- UK Dementia Research Institute, UCL Institute of NeurologyUniversity College LondonLondonUK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonUSA
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryUniversity of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalGothenburgSweden
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- Department of Psychiatry and NeurochemistryUniversity of GothenburgMölndalSweden
- Department of NeuropsychiatryRegion Västra GötalandSahlgrenska University HospitalGötalandSweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| |
Collapse
|
11
|
Cano A, Capdevila M, Puerta R, Arranz J, Montrreal L, de Rojas I, García-González P, Olivé C, García-Gutiérrez F, Sotolongo-Grau O, Orellana A, Aguilera N, Ramis M, Rosende-Roca M, Lleó A, Fortea J, Tartari JP, Lafuente A, Vargas L, Pérez-Cordón A, Muñoz N, Sanabria Á, Alegret M, Morató X, Tárraga L, Fernández V, Marquié M, Valero S, Alcolea D, Boada M, Ruiz A. Clinical value of plasma pTau181 to predict Alzheimer's disease pathology in a large real-world cohort of a memory clinic. EBioMedicine 2024; 108:105345. [PMID: 39299003 PMCID: PMC11424964 DOI: 10.1016/j.ebiom.2024.105345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 09/02/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND The identification of patients with an elevated risk of developing Alzheimer's disease (AD) dementia and eligible for the disease-modifying treatments (DMTs) in the earliest stages is one of the greatest challenges in the clinical practice. Plasma biomarkers has the potential to predict these issues, but further research is still needed to translate them to clinical practice. Here we evaluated the clinical applicability of plasma pTau181 as a predictive marker of AD pathology in a large real-world cohort of a memory clinic. METHODS Three independent cohorts (modelling [n = 991, 59.7% female], testing [n = 642, 56.2% female] and validation [n = 441, 55.1% female]) of real-world patients with subjective cognitive decline (SCD), mild cognitive impairment (MCI), AD dementia, and other dementias were included. Paired cerebrospinal fluid (CSF) and plasma samples were used to measure AT(N) CSF biomarkers and plasma pTau181. FINDINGS CSF and plasma pTau181 showed correlation in all phenotypes except in SCD and other dementias. Age significantly influenced the biomarker's performance. The general Aβ(+) vs Aβ(-) ROC curve showed an AUC = 0.77 [0.74-0.80], whereas the specific ROC curve of MCI due to AD vs non-AD MCI showed an AUC = 0.89 [0.85-0.93]. A cut-off value of 1.30 pg/ml of plasma pTau181 exhibited a sensitivity of 93.57% [88.72-96.52], specificity of 72.38% [62.51-79.01], VPP of 77.85% [70.61-83.54], and 8.30% false negatives in the subjects with MCI of the testing cohort. The HR of cox regression showed that patients with MCI up to this cut-off value exhibited a HR = 1.84 [1.05-3.22] higher risk to convert to AD dementia than patients with MCI below the cut-off value. INTERPRETATION Plasma pTau181 has the potential to be used in the memory clinics as a screening biomarker of AD pathology in subjects with MCI, presenting a valuable prognostic utility in predicting the MCI conversion to AD dementia. In the context of a real-world population, a confirmatory test employing gold-standard procedures is still advisable. FUNDING This study has been mainly funded by Ace Alzheimer Center Barcelona, Instituto de Salud Carlos III (ISCIII), Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Spanish Ministry of Science and Innovation, Fundación ADEY, Fundación Echevarne and Grífols S.A.
Collapse
Affiliation(s)
- Amanda Cano
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| | - María Capdevila
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain; Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Spain
| | - Raquel Puerta
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain
| | - Javier Arranz
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Laura Montrreal
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain
| | - Itziar de Rojas
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Pablo García-González
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Claudia Olivé
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain
| | | | - Oscar Sotolongo-Grau
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain
| | - Adelina Orellana
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Nuria Aguilera
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain
| | - Maribel Ramis
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain
| | - Maitee Rosende-Roca
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Alberto Lleó
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Juan Fortea
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autònoma de Barcelona, Barcelona, Spain; Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Juan Pablo Tartari
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain
| | - Asunción Lafuente
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain
| | - Liliana Vargas
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain
| | - Alba Pérez-Cordón
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain
| | - Nathalia Muñoz
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain
| | - Ángela Sanabria
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain
| | - Montserrat Alegret
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Xavier Morató
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain
| | - Lluís Tárraga
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Victoria Fernández
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain
| | - Marta Marquié
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Sergi Valero
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Daniel Alcolea
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mercè Boada
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Agustín Ruiz
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX, USA
| |
Collapse
|
12
|
Blum D, Cailliau E, Béhal H, Vidal J, Delaby C, Buée L, Allinquant B, Gabelle A, Bombois S, Lehmann S, Schraen‐Maschke S, Hanon O. Association of caffeine consumption with cerebrospinal fluid biomarkers in mild cognitive impairment and Alzheimer's disease: A BALTAZAR cohort study. Alzheimers Dement 2024; 20:6948-6959. [PMID: 39099181 PMCID: PMC11485411 DOI: 10.1002/alz.14169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/20/2024] [Accepted: 07/12/2024] [Indexed: 08/06/2024]
Abstract
INTRODUCTION We investigated the link between habitual caffeine intake with memory impairments and cerebrospinal fluid (CSF) biomarkers in mild cognitive impairment (MCI) and Alzheimer's disease (AD) patients. METHODS MCI (N = 147) and AD (N = 116) patients of the Biomarker of AmyLoid pepTide and AlZheimer's diseAse Risk (BALTAZAR) cohort reported their caffeine intake at inclusion using a dedicated survey. Associations of caffeine consumption with memory impairments and CSF biomarkers (tau, p-tau181, amyloid beta 1-42 [Aβ1-42], Aβ1-40) were analyzed using logistic and analysis of covariance models. RESULTS Adjusted on Apolipoprotein E (APOE ε4), age, sex, education level, and tobacco, lower caffeine consumption was associated with higher risk to be amnestic (OR: 2.49 [95% CI: 1.13 to 5.46]; p = 0.023) and lower CSF Aβ1-42 (p = 0.047), Aβ1-42/Aβ1-40 (p = 0.040), and Aβ1-42/p-tau181 (p = 0.020) in the whole cohort. DISCUSSION Data support the beneficial effect of caffeine consumption to memory impairments and CSF amyloid markers in MCI and AD patients. HIGHLIGHTS We studied the impact of caffeine consumption in the BALTAZAR cohort. Low caffeine intake is associated with higher risk of being amnestic in MCI/AD patients. Caffeine intake is associated with CSF biomarkers in AD patients.
Collapse
Affiliation(s)
- David Blum
- University of Lille, Inserm, CHU LilleUMR‐S1172 Lille Neuroscience & Cognition (LilNCog)LilleFrance
- Alzheimer and TauopathiesLabEx DISTALZLilleFrance
| | | | | | - Jean‐Sébastien Vidal
- Université Paris CitéINSERM U1144, GHU APHP CentreHopital Broca, Memory Resource and Research Centre de Paris‐Broca‐Ile de FranceParisFrance
| | - Constance Delaby
- Laboratoire et Plateforme de Protéomique CliniqueUniversité de MontpellierINM INSERM, IRMB CHU de Montpellier, 80 av FlicheMontpellierFrance
- Sant Pau Memory UnitHospital de la Santa Creu i Sant Pau ‐ Biomedical Research Institute Sant Pau ‐ Universitat Autònoma de BarcelonaBarcelonaSpain
| | - Luc Buée
- University of Lille, Inserm, CHU LilleUMR‐S1172 Lille Neuroscience & Cognition (LilNCog)LilleFrance
- Alzheimer and TauopathiesLabEx DISTALZLilleFrance
| | - Bernadette Allinquant
- Université Paris CitéInstitute of Psychiatry and Neuroscience, Inserm, UMR‐S 1266ParisFrance
| | - Audrey Gabelle
- Université de MontpellierCHU MontpellierMemory Research and Resources CenterDepartment of Neurology, Inserm INM NeuroPEPs TeamExcellence Center of Neurodegenerative DisordersMontpellierFrance
| | - Stéphanie Bombois
- Alzheimer and TauopathiesLabEx DISTALZLilleFrance
- Assistance Publique‐Hôpitaux de Paris (AP‐HP)Département de Neurologie, Centre des Maladies Cognitives et Comportementales, GH Pitié‐SalpêtrièreParisFrance
| | - Sylvain Lehmann
- Laboratoire et Plateforme de Protéomique CliniqueUniversité de MontpellierINM INSERM, IRMB CHU de Montpellier, 80 av FlicheMontpellierFrance
| | - Susanna Schraen‐Maschke
- University of Lille, Inserm, CHU LilleUMR‐S1172 Lille Neuroscience & Cognition (LilNCog)LilleFrance
- Alzheimer and TauopathiesLabEx DISTALZLilleFrance
| | - Olivier Hanon
- Université Paris CitéINSERM U1144, GHU APHP CentreHopital Broca, Memory Resource and Research Centre de Paris‐Broca‐Ile de FranceParisFrance
| |
Collapse
|
13
|
Ishiguro T, Kasuga K. Alzheimer's Disease-Related Cerebrospinal Fluid Biomarkers in Progressive Supranuclear Palsy. Brain Sci 2024; 14:859. [PMID: 39335355 PMCID: PMC11430815 DOI: 10.3390/brainsci14090859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/18/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
Progressive Supranuclear Palsy (PSP) is the most common four-repeat tauopathy. PSP cases are typically characterized by vertical gaze palsy and postural instability; however, various phenotypes have been reported, making antemortem diagnosis based on clinical symptoms challenging. The development of biomarkers reflecting brain pathology and the ability to diagnose patients based on these biomarkers are essential for developing future intervention strategies, including disease-modifying therapies. However, despite many dedicated efforts, no highly specific fluid biomarker for PSP has yet been established. Conversely, several cerebrospinal fluid (CSF) biomarkers of Alzheimer's Disease (AD) have been established, and an AT(N) classification system has been proposed. Typically, among patients with AD, CSF amyloid β42 (Aβ42), but not Aβ40, is decreased, resulting in a reduction in the Aβ42/Aβ40 ratio, while tau phosphorylated at threonine 181 (p-tau181) and total tau (t-tau) are increased. Interestingly, the core CSF AD biomarkers show unique patterns in patients with PSP. Furthermore, reports have indicated that the CSF levels of both Aβ42 and Aβ40 are decreased independently of Aβ accumulation in PSP. Therefore, the Aβ42/Aβ40 ratio could potentially be used to differentiate PSP from AD. Additionally, studies have reported that CSF p-tau and t-tau are reduced in PSP, and that the neurofilament light chain is remarkably increased compared to healthy controls and patients with AD, even though PSP is a neurodegenerative disease associated with tau accumulation. These PSP-specific changes in AD-related core biomarkers may reflect the pathology of PSP and contribute to its diagnosis. As such, elucidating the mechanisms underlying the observed decreases in Aβ and tau levels could facilitate a better understanding of the pathogenesis of PSP.
Collapse
Affiliation(s)
- Takanobu Ishiguro
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata 951-8585, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata 951-8585, Japan
| |
Collapse
|
14
|
Rajendrakumar AL, Arbeev KG, Bagley O, Yashin AI, Ukraintseva S. The association between rs6859 in NECTIN2 gene and Alzheimer's disease is partly mediated by pTau. Front Aging Neurosci 2024; 16:1388363. [PMID: 39165837 PMCID: PMC11334082 DOI: 10.3389/fnagi.2024.1388363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/18/2024] [Indexed: 08/22/2024] Open
Abstract
Introduction Emerging evidence suggests a connection between vulnerability to infections and Alzheimer's disease (AD). The nectin cell adhesion molecule 2 (NECTIN2) gene coding for a membrane component of adherens junctions is involved in response to infections, and its single nucleotide polymorphism (SNP) rs6859 was significantly associated with AD risk in several human cohorts. It is unclear, however, how exactly rs6859 influences the development of AD pathology. The aggregation of hyperphosphorylated tau protein (pTau) is a key pathological feature of neurodegeneration in AD, which may be induced by infections, among other factors, and potentially influenced by genes involved in both AD and vulnerability to infections, such as NECTIN2. Materials and methods We conducted a causal mediation analysis (CMA) on a sample of 708 participants in the Alzheimer's disease Neuroimaging Initiative (ADNI). The relationship between rs6859 and Alzheimer's disease (AD), with AD (yes/no) as the outcome and pTau-181 levels in the cerebrospinal fluid (CSF) acting as a mediator in this association, was assessed. Adjusted estimates from the probit and linear regression models were used in the CMA model, where an additive model considered an increase in dosage of the rs6859 A allele (AD risk factor). Results The increase in dose of allele A of the SNP rs6859 resulted in about 0.144 increase per standard deviation (SD) of pTau-181 (95% CI: 0.041, 0.248, p < 0.01). When included together in the probit model, the change in A allele dose and each standard deviation change in pTau-181 predicted 6.84% and 9.79% higher probabilities for AD, respectively. In the CMA, the proportion of the average mediated effect was 17.05% and was higher for the risk allele homozygotes (AA), at 19.40% (95% CI: 6.20%, 43.00%, p < 0.01). The sensitivity analysis confirmed the evidence of a robust mediation effect. Conclusion This study reported a new potential causal relationship between pTau-181 and AD. We found that the association between rs6859 in the NECTIN2 gene and AD is partly mediated by pTau-181 levels in CSF. The rest of this association may be mediated by other factors. Our finding sheds light on the complex interplay between genetic susceptibility, protein aggregation, and neurodegeneration in AD. Further research, using other biomarkers, is needed to uncover the remaining mechanisms of the association between the NECTIN2 gene and AD.
Collapse
Affiliation(s)
| | | | | | | | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| |
Collapse
|
15
|
Rajendrakumar AL, Arbeev KG, Bagley O, Yashin AI, Ukraintseva S. The association between rs6859 in NECTIN2 gene and Alzheimer's disease is partly mediated by pTau. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.21.24309310. [PMID: 38947013 PMCID: PMC11213054 DOI: 10.1101/2024.06.21.24309310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Introduction Emerging evidence suggests a connection between vulnerability to infections and Alzheimer's disease (AD). The nectin cell adhesion molecule 2 (NECTIN2) gene coding for a membrane component of adherens junctions is involved in response to infection, and its single nucleotide polymorphism (SNP) rs6859 was significantly associated with AD risk in several human cohorts. It is unclear, however, how exactly rs6859 influences the development of AD pathology. The aggregation of hyperphosphorylated tau protein (pTau) is a key pathological feature of neurodegeneration in AD, which may be induced by infections, among other factors, and potentially influenced by genes involved in both AD and vulnerability to infections, such as NECTIN2. Materials and methods We conducted a causal mediation analysis (CMA) on a sample of 708 participants in the Alzheimer's Disease Neuroimaging Initiative (ADNI). The relationship between rs6859 and Alzheimer's disease (AD), with AD (yes/no) as the outcome and pTau-181 levels in the cerebrospinal fluid (CSF) acting as a mediator in this association, was assessed. Adjusted estimates from the probit and linear regression models were used in the CMA model, where an additive model considered an increase in dosage of the rs6859 A allele (AD risk factor). Results The increase in dose of allele A of the SNP rs6859 resulted in about 0.144 increase per standard deviation (SD) of pTau-181 (95% CI: 0.041, 0.248, p<0.01). When included together in the probit model, the change in A allele dose and each standard deviation change in pTau-181 predicted 6.84% and 9.79% higher probabilities for AD, respectively. In the CMA, the proportion of the average mediated effect was 17.05% and was higher for the risk allele homozygotes (AA), at 19.40% (95% CI: 6.20%, 43.00%, p<0.01). The sensitivity analysis confirmed the evidence of a robust mediation effect. Conclusion This study reported a new causal relationship between pTau-181 and AD. We found that the association between rs6859 in the NECTIN2 gene and AD is partly mediated by pTau-181 levels in CSF. The rest of this association may be mediated by other factors. Further research, using other biomarkers, is needed to uncover the remaining mechanisms of the association between the NECTIN2 gene and AD.
Collapse
Affiliation(s)
| | - Konstantin G. Arbeev
- Biodemography of Aging Research Unit, Duke University, Social Science Research Institute, Durham, NC, USA
| | - Olivia Bagley
- Biodemography of Aging Research Unit, Duke University, Social Science Research Institute, Durham, NC, USA
| | - Anatoliy I. Yashin
- Biodemography of Aging Research Unit, Duke University, Social Science Research Institute, Durham, NC, USA
| | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Duke University, Social Science Research Institute, Durham, NC, USA
| |
Collapse
|
16
|
Lohman T, Kapoor A, Engstrom AC, Shenasa F, Alitin JPM, Gaubert A, Rodgers KE, Bradford D, Mather M, Han SD, Head E, Sordo L, Thayer JF, Nation DA. Central autonomic network dysfunction and plasma Alzheimer's disease biomarkers in older adults. Alzheimers Res Ther 2024; 16:124. [PMID: 38851772 PMCID: PMC11162037 DOI: 10.1186/s13195-024-01486-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/24/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Higher order regulation of autonomic function is maintained by the coordinated activity of specific cortical and subcortical brain regions, collectively referred to as the central autonomic network (CAN). Autonomic changes are frequently observed in Alzheimer's disease (AD) and dementia, but no studies to date have investigated whether plasma AD biomarkers are associated with CAN functional connectivity changes in at risk older adults. METHODS Independently living older adults (N = 122) without major neurological or psychiatric disorder were recruited from the community. Participants underwent resting-state brain fMRI and a CAN network derived from a voxel-based meta-analysis was applied for overall, sympathetic, and parasympathetic CAN connectivity using the CONN Functional Toolbox. Sensorimotor network connectivity was studied as a negative control. Plasma levels of amyloid (Aβ42, Aβ40), neurofilament light chain (NfL), and glial fibrillary acidic protein (GFAP) were assessed using digital immunoassay. The relationship between plasma AD biomarkers and within-network functional connectivity was studied using multiple linear regression adjusted for demographic covariates and Apolipoprotein E (APOE) genotype. Interactive effects with APOE4 carrier status were also assessed. RESULTS All autonomic networks were positively associated with Aβ42/40 ratio and remained so after adjustment for age, sex, and APOE4 carrier status. Overall and parasympathetic networks were negatively associated with GFAP. The relationship between the parasympathetic CAN and GFAP was moderated by APOE4 carrier status, wherein APOE4 carriers with low parasympathetic CAN connectivity displayed the highest plasma GFAP concentrations (B = 910.00, P = .004). Sensorimotor connectivity was not associated with any plasma AD biomarkers, as expected. CONCLUSION The present study findings suggest that CAN function is associated with plasma AD biomarker levels. Specifically, lower CAN functional connectivity is associated with decreased plasma Aβ42/40, indicative of cerebral amyloidosis, and increased plasma GFAP in APOE4 carriers at risk for AD. These findings could suggest higher order autonomic and parasympathetic dysfunction in very early-stage AD, which may have clinical implications.
Collapse
Affiliation(s)
- Trevor Lohman
- University of Southern California, Leonard Davis School of Gerontology, Los Angeles, CA, USA
| | - Arunima Kapoor
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Allison C Engstrom
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Fatemah Shenasa
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - John Paul M Alitin
- University of Southern California, Leonard Davis School of Gerontology, Los Angeles, CA, USA
| | - Aimee Gaubert
- University of Southern California, Leonard Davis School of Gerontology, Los Angeles, CA, USA
| | - Kathleen E Rodgers
- Center for Innovations in Brain Science, Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - David Bradford
- Center for Innovations in Brain Science, Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Mara Mather
- University of Southern California, Leonard Davis School of Gerontology, Los Angeles, CA, USA
| | - S Duke Han
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | - Lorena Sordo
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | - Julian F Thayer
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Daniel A Nation
- University of Southern California, Leonard Davis School of Gerontology, Los Angeles, CA, USA.
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
17
|
Wang YT, Therriault J, Servaes S, Tissot C, Rahmouni N, Macedo AC, Fernandez-Arias J, Mathotaarachchi SS, Benedet AL, Stevenson J, Ashton NJ, Lussier FZ, Pascoal TA, Zetterberg H, Rajah MN, Blennow K, Gauthier S, Rosa-Neto P. Sex-specific modulation of amyloid-β on tau phosphorylation underlies faster tangle accumulation in females. Brain 2024; 147:1497-1510. [PMID: 37988283 PMCID: PMC10994548 DOI: 10.1093/brain/awad397] [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: 08/10/2023] [Revised: 10/26/2023] [Accepted: 10/28/2023] [Indexed: 11/23/2023] Open
Abstract
Females are disproportionately affected by dementia due to Alzheimer's disease. Despite a similar amyloid-β (Aβ) load, a higher load of neurofibrillary tangles (NFTs) is seen in females than males. Previous literature has proposed that Aβ and phosphorylated-tau (p-tau) synergism accelerates tau tangle formation, yet the effect of biological sex in this process has been overlooked. In this observational study, we examined longitudinal neuroimaging data from the TRIAD and ADNI cohorts from Canada and USA, respectively. We assessed 457 participants across the clinical spectrum of Alzheimer's disease. All participants underwent baseline multimodal imaging assessment, including MRI and PET, with radioligands targeting Aβ plaques and tau tangles, respectively. CSF data were also collected. Follow-up imaging assessments were conducted at 1- and 2-year intervals for the TRIAD cohort and 1-, 2- and 4-year intervals for the ADNI cohort. The upstream pathological events contributing to faster tau progression in females were investigated-specifically, whether the contribution of Aβ and p-tau synergism to accelerated tau tangle formation is modulated by biological sex. We hypothesized that cortical Aβ predisposes tau phosphorylation and tangle accumulation in a sex-specific manner. Findings revealed that Aβ-positive females presented higher CSF p-tau181 concentrations compared with Aβ-positive males in both the TRIAD (P = 0.04, Cohen's d = 0.51) and ADNI (P = 0.027, Cohen's d = 0.41) cohorts. In addition, Aβ-positive females presented faster NFT accumulation compared with their male counterparts (TRIAD: P = 0.026, Cohen's d = 0.52; ADNI: P = 0.049, Cohen's d = 1.14). Finally, the triple interaction between female sex, Aβ and CSF p-tau181 was revealed as a significant predictor of accelerated tau accumulation at the 2-year follow-up visit (Braak I: P = 0.0067, t = 2.81; Braak III: P = 0.017, t = 2.45; Braak IV: P = 0.002, t = 3.17; Braak V: P = 0.006, t = 2.88; Braak VI: P = 0.0049, t = 2.93). Overall, we report sex-specific modulation of cortical Aβ in tau phosphorylation, consequently facilitating faster NFT progression in female individuals over time. This presents important clinical implications and suggests that early intervention that targets Aβ plaques and tau phosphorylation may be a promising therapeutic strategy in females to prevent the further accumulation and spread of tau aggregates.
Collapse
Affiliation(s)
- Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Arthur Cassa Macedo
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Jaime Fernandez-Arias
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Sulantha S Mathotaarachchi
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Andréa L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 41 Mölndal, Sweden
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 41 Mölndal, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, 4011 Stavanger, Norway
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London SE5 9RX, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London SE5 8AF, UK
| | - Firoza Z Lussier
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Tharick A Pascoal
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 41 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
- UK Dementia Research Institute at UCL, London WC1E 6BT, 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, University of Wisconsin-Madison, Madison, WI 53792, USA
| | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 41 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
- Montreal Neurological Institute, Montreal, QC H3A 2B4, Canada
| |
Collapse
|
18
|
Colvee-Martin H, Parra JR, Gonzalez GA, Barker W, Duara R. Neuropathology, Neuroimaging, and Fluid Biomarkers in Alzheimer's Disease. Diagnostics (Basel) 2024; 14:704. [PMID: 38611617 PMCID: PMC11012058 DOI: 10.3390/diagnostics14070704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/05/2024] [Accepted: 02/17/2024] [Indexed: 04/14/2024] Open
Abstract
An improved understanding of the pathobiology of Alzheimer's disease (AD) should lead ultimately to an earlier and more accurate diagnosis of AD, providing the opportunity to intervene earlier in the disease process and to improve outcomes. The known hallmarks of Alzheimer's disease include amyloid-β plaques and neurofibrillary tau tangles. It is now clear that an imbalance between production and clearance of the amyloid beta protein and related Aβ peptides, especially Aβ42, is a very early, initiating factor in Alzheimer's disease (AD) pathogenesis, leading to aggregates of hyperphosphorylation and misfolded tau protein, inflammation, and neurodegeneration. In this article, we review how the AD diagnostic process has been transformed in recent decades by our ability to measure these various elements of the pathological cascade through the use of imaging and fluid biomarkers. The more recently developed plasma biomarkers, especially phosphorylated-tau217 (p-tau217), have utility for screening and diagnosis of the earliest stages of AD. These biomarkers can also be used to measure target engagement by disease-modifying therapies and the response to treatment.
Collapse
Affiliation(s)
- Helena Colvee-Martin
- Wien Center for Alzheimer’s Disease & Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (H.C.-M.); (W.B.)
| | - Juan Rayo Parra
- Human & Molecular Genetics, Florida International University, Miami, FL 33199, USA; (J.R.P.); (G.A.G.)
| | - Gabriel Antonio Gonzalez
- Human & Molecular Genetics, Florida International University, Miami, FL 33199, USA; (J.R.P.); (G.A.G.)
| | - Warren Barker
- Wien Center for Alzheimer’s Disease & Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (H.C.-M.); (W.B.)
| | - Ranjan Duara
- Wien Center for Alzheimer’s Disease & Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (H.C.-M.); (W.B.)
| |
Collapse
|
19
|
Levin F, Grothe MJ, Dyrba M, Franzmeier N, Teipel SJ. Longitudinal trajectories of cognitive reserve in hypometabolic subtypes of Alzheimer's disease. Neurobiol Aging 2024; 135:26-38. [PMID: 38157587 DOI: 10.1016/j.neurobiolaging.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 11/16/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024]
Abstract
Previous studies have demonstrated resilience to AD-related neuropathology in a form of cognitive reserve (CR). In this study we investigated a relationship between CR and hypometabolic subtypes of AD, specifically the typical and the limbic-predominant subtypes. We analyzed data from 59 Aβ-positive cognitively normal (CN), 221 prodromal Alzheimer's disease (AD) and 174 AD dementia participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI) from ADNI and ADNIGO/2 phases. For replication, we analyzed data from 5 Aβ-positive CN, 89 prodromal AD and 43 AD dementia participants from ADNI3. CR was estimated as standardized residuals in a model predicting cognition from temporoparietal grey matter volumes and covariates. Higher CR estimates predicted slower cognitive decline. Typical and limbic-predominant hypometabolic subtypes demonstrated similar baseline CR, but the results suggested a faster decline of CR in the typical subtype. These findings support the relationship between subtypes and CR, specifically longitudinal trajectories of CR. Results also underline the importance of longitudinal analyses in research on CR.
Collapse
Affiliation(s)
- Fedor Levin
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Rostock, Germany.
| | - Michel J Grothe
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Martin Dyrba
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Rostock, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Sweden; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Stefan J Teipel
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Rostock, Germany; Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
20
|
Chapleau M, La Joie R, Yong K, Agosta F, Allen IE, Apostolova L, Best J, Boon BDC, Crutch S, Filippi M, Fumagalli GG, Galimberti D, Graff-Radford J, Grinberg LT, Irwin DJ, Josephs KA, Mendez MF, Mendez PC, Migliaccio R, Miller ZA, Montembeault M, Murray ME, Nemes S, Pelak V, Perani D, Phillips J, Pijnenburg Y, Rogalski E, Schott JM, Seeley W, Sullivan AC, Spina S, Tanner J, Walker J, Whitwell JL, Wolk DA, Ossenkoppele R, Rabinovici GD. Demographic, clinical, biomarker, and neuropathological correlates of posterior cortical atrophy: an international cohort study and individual participant data meta-analysis. Lancet Neurol 2024; 23:168-177. [PMID: 38267189 PMCID: PMC11615965 DOI: 10.1016/s1474-4422(23)00414-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/22/2023] [Accepted: 10/18/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND Posterior cortical atrophy is a rare syndrome characterised by early, prominent, and progressive impairment in visuoperceptual and visuospatial processing. The disorder has been associated with underlying neuropathological features of Alzheimer's disease, but large-scale biomarker and neuropathological studies are scarce. We aimed to describe demographic, clinical, biomarker, and neuropathological correlates of posterior cortical atrophy in a large international cohort. METHODS We searched PubMed between database inception and Aug 1, 2021, for all published research studies on posterior cortical atrophy and related terms. We identified research centres from these studies and requested deidentified, individual participant data (published and unpublished) that had been obtained at the first diagnostic visit from the corresponding authors of the studies or heads of the research centres. Inclusion criteria were a clinical diagnosis of posterior cortical atrophy as defined by the local centre and availability of Alzheimer's disease biomarkers (PET or CSF), or a diagnosis made at autopsy. Not all individuals with posterior cortical atrophy fulfilled consensus criteria, being diagnosed using centre-specific procedures or before development of consensus criteria. We obtained demographic, clinical, biofluid, neuroimaging, and neuropathological data. Mean values for continuous variables were combined using the inverse variance meta-analysis method; only research centres with more than one participant for a variable were included. Pooled proportions were calculated for binary variables using a restricted maximum likelihood model. Heterogeneity was quantified using I2. FINDINGS We identified 55 research centres from 1353 papers, with 29 centres responding to our request. An additional seven centres were recruited by advertising via the Alzheimer's Association. We obtained data for 1092 individuals who were evaluated at 36 research centres in 16 countries, the other sites having not responded to our initial invitation to participate to the study. Mean age at symptom onset was 59·4 years (95% CI 58·9-59·8; I2=77%), 60% (56-64; I2=35%) were women, and 80% (72-89; I2=98%) presented with posterior cortical atrophy pure syndrome. Amyloid β in CSF (536 participants from 28 centres) was positive in 81% (95% CI 75-87; I2=78%), whereas phosphorylated tau in CSF (503 participants from 29 centres) was positive in 65% (56-75; I2=87%). Amyloid-PET (299 participants from 24 centres) was positive in 94% (95% CI 90-97; I2=15%), whereas tau-PET (170 participants from 13 centres) was positive in 97% (93-100; I2=12%). At autopsy (145 participants from 13 centres), the most frequent neuropathological diagnosis was Alzheimer's disease (94%, 95% CI 90-97; I2=0%), with common co-pathologies of cerebral amyloid angiopathy (71%, 54-88; I2=89%), Lewy body disease (44%, 25-62; I2=77%), and cerebrovascular injury (42%, 24-60; I2=88%). INTERPRETATION These data indicate that posterior cortical atrophy typically presents as a pure, young-onset dementia syndrome that is highly specific for underlying Alzheimer's disease pathology. Further work is needed to understand what drives cognitive vulnerability and progression rates by investigating the contribution of sex, genetics, premorbid cognitive strengths and weaknesses, and brain network integrity. FUNDING None.
Collapse
Affiliation(s)
- Marianne Chapleau
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Keir Yong
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London, UK
| | - Federica Agosta
- Vita-Salute, San Raffaele University, Milan, Italy; Neuroimaging Research Unit, Division of Neuroscience, and Neurology Unit, IRCCS San Raffaele Scientific Insitute, Milan, Italy
| | - Isabel Elaine Allen
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | | | - John Best
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Baayla D C Boon
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Sebastian Crutch
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London, UK
| | - Massimo Filippi
- Vita-Salute, San Raffaele University, Milan, Italy; Neuroimaging Research Unit, Division of Neuroscience, and Neurology Unit, IRCCS San Raffaele Scientific Insitute, Milan, Italy
| | | | - Daniela Galimberti
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy; Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | | | - Lea T Grinberg
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Department of Pathology, University of California San Francisco, San Francisco, CA, USA; Department of Pathology, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - David J Irwin
- Penn Frontotemporal Degeneration Center, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Mario F Mendez
- David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Patricio Chrem Mendez
- Memory Center, Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia, Buenos Aires Argentina
| | - Raffaella Migliaccio
- Paris Brain Institute (ICM), FrontLab, Institut de la mémoire et de la maladie d'Alzheimer (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, Paris, France
| | - Zachary A Miller
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Maxime Montembeault
- Douglas Research Centre, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | | | - Sára Nemes
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Victoria Pelak
- Departments of Neurology and Ophthalmology, Divisions of Neuro-Ophthalmology and Behavioral Neurology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Daniela Perani
- Vita-Salute, San Raffaele University, Milan, Italy; Division of Neuroscience, IRCCS San Raffaele, San Raffaele University, Milan, Italy
| | - Jeffrey Phillips
- Penn Frontotemporal Degeneration Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Yolande Pijnenburg
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, Netherlands; Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands
| | - Emily Rogalski
- Mesulam Center for Cognitive Neurology & Alzheimer's Disease, Northwestern University, Evanston, IL, USA
| | - Jonathan M Schott
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London, UK; Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, Netherlands
| | - William Seeley
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - A Campbell Sullivan
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Salvatore Spina
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Jeremy Tanner
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Jamie Walker
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | | | - David A Wolk
- Alzheimer's Disease Research Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, Netherlands; Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands; Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Department of Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
21
|
Muir RT, Ismail Z, Black SE, Smith EE. Comparative methods for quantifying plasma biomarkers in Alzheimer's disease: Implications for the next frontier in cerebral amyloid angiopathy diagnostics. Alzheimers Dement 2024; 20:1436-1458. [PMID: 37908054 PMCID: PMC10916950 DOI: 10.1002/alz.13510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 08/09/2023] [Accepted: 08/13/2023] [Indexed: 11/02/2023]
Abstract
Plasma amyloid beta (Aβ) and tau are emerging as accessible biomarkers for Alzheimer's disease (AD). However, many assays exist with variable test performances, highlighting the need for a comparative assessment to identify the most valid assays for future use in AD and to apply to other settings in which the same biomarkers may be useful, namely, cerebral amyloid angiopathy (CAA). CAA is a progressive cerebrovascular disease characterized by deposition of Aβ40 and Aβ42 in cortical and leptomeningeal vessels. Novel immunotherapies for AD can induce amyloid-related imaging abnormalities resembling CAA-related inflammation. Few studies have evaluated plasma biomarkers in CAA. Identifying a CAA signature could facilitate diagnosis, prognosis, and a safer selection of patients with AD for emerging immunotherapies. This review evaluates studies that compare the diagnostic test performance of plasma biomarker techniques in AD and cerebrovascular and plasma biomarker profiles of CAA; it also discusses novel hypotheses and future avenues for plasma biomarker research in CAA.
Collapse
Affiliation(s)
- Ryan T. Muir
- Calgary Stroke ProgramDepartment of Clinical NeurosciencesUniversity of CalgaryCalgaryAlbertaCanada
- Department of Community Health SciencesUniversity of CalgaryCalgaryAlbertaCanada
- Hotchkiss Brain InstituteUniversity of CalgaryCalgaryAlbertaCanada
| | - Zahinoor Ismail
- Department of Community Health SciencesUniversity of CalgaryCalgaryAlbertaCanada
- Hotchkiss Brain InstituteUniversity of CalgaryCalgaryAlbertaCanada
- Department of PsychiatryUniversity of CalgaryCalgaryAlbertaCanada
| | - Sandra E. Black
- Division of NeurologyDepartment of MedicineSunnybrook Health Sciences CentreTorontoOntarioCanada
- LC Campbell Cognitive Neurology Research UnitDr Sandra Black Centre for Brain Resilience and Recovery, and Hurvitz Brain Sciences ProgramSunnybrook Research InstituteUniversity of TorontoTorontoOntarioCanada
| | - Eric E. Smith
- Calgary Stroke ProgramDepartment of Clinical NeurosciencesUniversity of CalgaryCalgaryAlbertaCanada
- Department of Community Health SciencesUniversity of CalgaryCalgaryAlbertaCanada
- Hotchkiss Brain InstituteUniversity of CalgaryCalgaryAlbertaCanada
| |
Collapse
|
22
|
Kimura T, Sato H, Kano M, Tatsumi L, Tomita T. Novel aspects of the phosphorylation and structure of pathological tau: implications for tauopathy biomarkers. FEBS Open Bio 2024; 14:181-193. [PMID: 37391389 PMCID: PMC10839341 DOI: 10.1002/2211-5463.13667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/17/2023] [Accepted: 06/29/2023] [Indexed: 07/02/2023] Open
Abstract
The deposition of highly phosphorylated and aggregated tau is a characteristic of tauopathies, including Alzheimer's disease. It has long been known that different isoforms of tau are aggregated in different cell types and brain regions in each tauopathy. Recent advances in analytical techniques revealed the details of the biochemical and structural biological differences of tau specific to each tauopathy. In this review, we explain recent advances in the analysis of post-translational modifications of tau, particularly phosphorylation, brought about by the development of mass-spectrometry and Phos-tag technology. We then discuss the structure of tau filaments in each tauopathy revealed by the advent of cryo-EM. Finally, we describe the progress in biofluid and imaging biomarkers for tauopathy. This review summarizes current efforts to elucidate the characteristics of pathological tau and the landscape of the use of tau as a biomarker to diagnose and determine the pathological stage of tauopathy.
Collapse
Affiliation(s)
- Taeko Kimura
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical SciencesThe University of TokyoJapan
| | - Haruaki Sato
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical SciencesThe University of TokyoJapan
| | - Maria Kano
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical SciencesThe University of TokyoJapan
| | - Lisa Tatsumi
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical SciencesThe University of TokyoJapan
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical SciencesThe University of TokyoJapan
| |
Collapse
|
23
|
Korologou-Linden R, Kalsi J, Kafetsouli D, Olawale A, Iwata A, Wingfield D, Mummery D, Hayhoe B, Robinson O, Majeed A, Middleton LT. Novel Blood-Based Biomarkers and Disease Modifying Therapies for Alzheimer's Disease. Are We Ready for the New Era? J Prev Alzheimers Dis 2024; 11:897-902. [PMID: 39044500 PMCID: PMC11266440 DOI: 10.14283/jpad.2024.83] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 03/17/2024] [Indexed: 07/25/2024]
Abstract
Recent positive trials for novel disease modifying therapies of anti-amyloid monoclonal antibodies represent a paradigm shift in the prevention and management of Alzheimer's disease, a relentlessly progressive and debilitating disease of old age. The reported efficacy of these new agents when given early in the disease trajectory is dependent on an early and accurate disease diagnosis, which is currently based on cerebrospinal fluid tests or/and neuro-imaging studies such as positron emission tomography. These confirmatory tests provide in vivo evidence of the pathological signature of Alzheimer's disease, of increased cerebral amyloid and tau burden and neurodegeneration. The emergence of blood-based biomarkers represents another breakthrough, offering a less invasive and scalable diagnostic tool that could be applied in both primary and specialist care settings, potentially revolutionizing Alzheimer's disease clinical pathways. However, healthcare systems face challenges in the adoption of these new technologies and therapies due to diagnostic and treatment capacity constraints, as well as financial and infrastructure requirements.
Collapse
Affiliation(s)
- R Korologou-Linden
- Roxanna Korologou-Linden, Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, 11th Floor, Charing Cross Hospital Campus, W6 8RP, , Tel: +44 20 3311 0208
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Rehman H, Ang TFA, Tao Q, Espenilla AL, Au R, Farrer LA, Zhang X, Qiu WQ. Comparison of Commonly Measured Plasma and Cerebrospinal Fluid Proteins and Their Significance for the Characterization of Cognitive Impairment Status. J Alzheimers Dis 2024; 97:621-633. [PMID: 38143358 DOI: 10.3233/jad-230837] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2023]
Abstract
BACKGROUND Although cerebrospinal fluid (CSF) amyloid-β42 peptide (Aβ42) and phosphorylated tau (p-tau) and blood p-tau are valuable for differential diagnosis of Alzheimer's disease (AD) from cognitively normal (CN) there is a lack of validated biomarkers for mild cognitive impairment (MCI). OBJECTIVE This study sought to determine how plasma and CSF protein markers compared in the characterization of MCI and AD status. METHODS This cohort study included Alzheimer's Disease Neuroimaging Initiative (ADNI) participants who had baseline levels of 75 proteins measured commonly in plasma and CSF (257 total, 46 CN, 143 MCI, and 68 AD). Logistic regression, least absolute shrinkage and selection operator (LASSO) and Random Forest (RF) methods were used to identify the protein candidates for the disease classification. RESULTS We observed that six plasma proteins panel (APOE, AMBP, C3, IL16, IGFBP2, APOD) outperformed the seven CSF proteins panel (VEGFA, HGF, PRL, FABP3, FGF4, CD40, RETN) as well as AD markers (CSF p-tau and Aβ42) to distinguish the MCI from AD [area under the curve (AUC) = 0.75 (plasma proteins), AUC = 0.60 (CSF proteins) and AUC = 0.56 (CSF p-tau and Aβ42)]. Also, these six plasma proteins performed better than the CSF proteins and were in line with CSF p-tau and Aβ42 in differentiating CN versus MCI subjects [AUC = 0.89 (plasma proteins), AUC = 0.85 (CSF proteins) and AUC = 0.89 (CSF p-tau and Aβ42)]. These results were adjusted for age, sex, education, and APOEϵ4 genotype. CONCLUSIONS This study suggests that the combination of 6 plasma proteins can serve as an effective marker for differentiating MCI from AD and CN.
Collapse
Affiliation(s)
- Habbiburr Rehman
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ting Fang Alvin Ang
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Qiushan Tao
- Department of Pharmacology & Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Arielle Lauren Espenilla
- Department of Biostatistics and Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Rhoda Au
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Boston University School of Medicine, Framingham, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Lindsay A Farrer
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics and Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Boston University School of Medicine, Framingham, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Xiaoling Zhang
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics and Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Boston University School of Medicine, Framingham, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Wei Qiao Qiu
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pharmacology & Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
25
|
Veitch DP, Weiner MW, Miller M, Aisen PS, Ashford MA, Beckett LA, Green RC, Harvey D, Jack CR, Jagust W, Landau SM, Morris JC, Nho KT, Nosheny R, Okonkwo O, Perrin RJ, Petersen RC, Rivera Mindt M, Saykin A, Shaw LM, Toga AW, Tosun D. The Alzheimer's Disease Neuroimaging Initiative in the era of Alzheimer's disease treatment: A review of ADNI studies from 2021 to 2022. Alzheimers Dement 2024; 20:652-694. [PMID: 37698424 PMCID: PMC10841343 DOI: 10.1002/alz.13449] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 09/13/2023]
Abstract
The Alzheimer's Disease Neuroimaging Initiative (ADNI) aims to improve Alzheimer's disease (AD) clinical trials. Since 2006, ADNI has shared clinical, neuroimaging, and cognitive data, and biofluid samples. We used conventional search methods to identify 1459 publications from 2021 to 2022 using ADNI data/samples and reviewed 291 impactful studies. This review details how ADNI studies improved disease progression understanding and clinical trial efficiency. Advances in subject selection, detection of treatment effects, harmonization, and modeling improved clinical trials and plasma biomarkers like phosphorylated tau showed promise for clinical use. Biomarkers of amyloid beta, tau, neurodegeneration, inflammation, and others were prognostic with individualized prediction algorithms available online. Studies supported the amyloid cascade, emphasized the importance of neuroinflammation, and detailed widespread heterogeneity in disease, linked to genetic and vascular risk, co-pathologies, sex, and resilience. Biological subtypes were consistently observed. Generalizability of ADNI results is limited by lack of cohort diversity, an issue ADNI-4 aims to address by enrolling a diverse cohort.
Collapse
Affiliation(s)
- Dallas P. Veitch
- Department of Veterans Affairs Medical CenterNorthern California Institute for Research and Education (NCIRE)San FranciscoCaliforniaUSA
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
| | - Michael W. Weiner
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of MedicineUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of Psychiatry and Behavioral SciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Melanie Miller
- Department of Veterans Affairs Medical CenterNorthern California Institute for Research and Education (NCIRE)San FranciscoCaliforniaUSA
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
| | - Paul S. Aisen
- Alzheimer's Therapeutic Research InstituteUniversity of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Miriam A. Ashford
- Department of Veterans Affairs Medical CenterNorthern California Institute for Research and Education (NCIRE)San FranciscoCaliforniaUSA
| | - Laurel A. Beckett
- Division of BiostatisticsDepartment of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | - Robert C. Green
- Division of GeneticsDepartment of MedicineBrigham and Women's HospitalBroad Institute Ariadne Labs and Harvard Medical SchoolBostonMassachusettsUSA
| | - Danielle Harvey
- Division of BiostatisticsDepartment of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | | | - William Jagust
- Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Susan M. Landau
- Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - John C. Morris
- Knight Alzheimer's Disease Research CenterWashington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
- Department of Pathology and ImmunologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Kwangsik T. Nho
- Department of Radiology and Imaging Sciences and the Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for Computational Biology and BioinformaticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Rachel Nosheny
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Psychiatry and Behavioral SciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Ozioma Okonkwo
- Wisconsin Alzheimer's Disease Research Center and Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Richard J. Perrin
- Knight Alzheimer's Disease Research CenterWashington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
- Department of Pathology and ImmunologyWashington University School of MedicineSaint LouisMissouriUSA
| | | | - Monica Rivera Mindt
- Department of PsychologyLatin American and Latino Studies InstituteAfrican and African American StudiesFordham UniversityNew YorkNew YorkUSA
- Department of NeurologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Andrew Saykin
- Department of Radiology and Imaging Sciences and the Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine and the PENN Alzheimer's Disease Research CenterCenter for Neurodegenerative ResearchPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Arthur W. Toga
- Laboratory of Neuro ImagingInstitute of Neuroimaging and InformaticsKeck School of Medicine of University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Duygu Tosun
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | | |
Collapse
|
26
|
Starmans NLP, Kappelle LJ, Muller M, Staals J, Teunissen CE, Biessels GJ, van der Flier WM, Wolters FJ. Blood Pressure Variability and Plasma Biomarkers of Neuronal Injury and Alzheimer's Disease: A Clinic-Based Study of Patients with Diseases Along the Heart-Brain Axis. J Alzheimers Dis 2024; 99:1207-1215. [PMID: 38788076 PMCID: PMC11191465 DOI: 10.3233/jad-240119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2024] [Indexed: 05/26/2024]
Abstract
Higher blood pressure variability (BPV) predisposes to cognitive decline. To investigate underlying mechanisms, we measured 24-h ambulatory BPV, nocturnal dipping and orthostatic hypotension in 518 participants with vascular cognitive impairment, carotid occlusive disease, heart failure, or reference participants. We determined cross-sectional associations between BPV indices and plasma biomarkers of neuronal injury (neurofilament light chain) and Alzheimer's disease (phosphorylated-tau-181 and Aβ42/Aβ40). None of the BPV indices were significantly associated with any of the biomarkers. Hence, in patients with diseases along the heart-brain axis, we found no evidence for an association between BPV and selected markers of neuronal injury or Alzheimer's disease.
Collapse
Affiliation(s)
| | - Laurens Jaap Kappelle
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Majon Muller
- Department of Internal Medicine, Geriatrics Section, Amsterdam Cardiovascular Science, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
| | - Julie Staals
- Department of Neurology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Charlotte Elisabeth Teunissen
- Department of Clinical Chemistry, Neurochemistry Laboratory, Amsterdam Neuroscience, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
| | - Geert Jan Biessels
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wiesje Maria van der Flier
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
- Department of Epidemiology, Amsterdam University Medical Center (Amsterdam UMC), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Frank Johannes Wolters
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine and Alzheimer Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - on behalf of the Heart-Brain Connection Consortium
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Internal Medicine, Geriatrics Section, Amsterdam Cardiovascular Science, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
- Department of Neurology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Clinical Chemistry, Neurochemistry Laboratory, Amsterdam Neuroscience, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
- Department of Epidemiology, Amsterdam University Medical Center (Amsterdam UMC), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine and Alzheimer Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
27
|
Nallapu BT, Petersen KK, Lipton RB, Davatzikos C, Ezzati A. Plasma Biomarkers as Predictors of Progression to Dementia in Individuals with Mild Cognitive Impairment. J Alzheimers Dis 2024; 98:231-246. [PMID: 38393899 PMCID: PMC11044769 DOI: 10.3233/jad-230620] [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: 02/25/2024]
Abstract
Background Blood-based biomarkers (BBMs) are of growing interest in the field of Alzheimer's disease (AD) and related dementias. Objective This study aimed to assess the ability of plasma biomarkers to 1) predict disease progression from mild cognitive impairment (MCI) to dementia and 2) improve the predictive ability of magnetic resonance imaging (MRI) and cerebrospinal fluid (CSF) measures when combined. Methods We used data from the Alzheimer's Disease Neuroimaging Initiative. Machine learning models were trained using the data from participants who remained cognitively stable (CN-s) and with Dementia diagnosis at 2-year follow-up visit. The models were used to predict progression to dementia in MCI individuals. We assessed the performance of models with plasma biomarkers against those with CSF and MRI measures, and also in combination with them. Results Our models with plasma biomarkers classified CN-s individuals from AD with an AUC of 0.75±0.03 and could predict conversion to dementia in MCI individuals with an AUC of 0.64±0.03 (17.1% BP, base prevalence). Models with plasma biomarkers performed better when combined with CSF and MRI measures (CN versus AD: AUC of 0.89±0.02; MCI-to-AD: AUC of 0.76±0.03, 21.5% BP). Conclusions Our results highlight the potential of plasma biomarkers in predicting conversion to dementia in MCI individuals. While plasma biomarkers could improve the predictive ability of CSF and MRI measures when combined, they also show the potential to predict non-progression to AD when considered alone. The predictive ability of plasma biomarkers is crucially linked to reducing the costly and effortful collection of CSF and MRI measures.
Collapse
Affiliation(s)
- Bhargav T. Nallapu
- Saul B. Korey Department of Neurology, Albert Einstein College of Medicine, New York City, NY, USA
| | - Kellen K. Petersen
- Saul B. Korey Department of Neurology, Albert Einstein College of Medicine, New York City, NY, USA
| | - Richard B. Lipton
- Saul B. Korey Department of Neurology, Albert Einstein College of Medicine, New York City, NY, USA
| | - Christos Davatzikos
- Radiology Department, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ali Ezzati
- Saul B. Korey Department of Neurology, Albert Einstein College of Medicine, New York City, NY, USA
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | | |
Collapse
|
28
|
Gonzalez-Ortiz F, Dias A, Turton M, Magalhães R, Kac PR, Correia M, Harrison P, Zetterberg H, Maia LF, Blennow K, Karikari TK. Preanalytical stability of plasma/serum brain-derived tau. Alzheimers Dement 2023; 19:4764-4770. [PMID: 37232524 PMCID: PMC10592602 DOI: 10.1002/alz.13156] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/27/2023]
Abstract
INTRODUCTION We investigated the effects of matrix type and reagent batch changes on diagnostic performances and longitudinal trajectories of brain-derived tau (BD-tau). METHODS We evaluated (i) Cohort 1: paired EDTA plasma and serum from Alzheimer biomarker-positive older adults versus controls (n = 26); and (ii) Cohort 2: n = 79 acute ischemic stroke patients with 265 longitudinal samples across four time points. RESULTS In Cohort 1, plasma and serum BD-tau were strongly correlated (rho = 0.96, p < 0.0001) with similar diagnostic performances (AUCs >99%) and correlations with CSF total-tau (rho = 0.93-0.94, p < 0.0001). However, absolute concentrations were ∼40% higher in plasma versus serum. In Cohort 2, first and repeated BD-tau measurements showed a near-perfect correlation (rho = 0.96, p < 0.0001), with no significant between-batch concentration differences. In longitudinal analyses, substituting ∼10% of the first-run concentrations for the remeasured values showed overlapping estimated trajectories without significant differences at any time point. DISCUSSION BD-tau has equivalent diagnostic accuracies, but non-interchangeable absolute concentrations, in plasma versus serum. Furthermore, the analytical robustness is unaffected by batch-to-batch reagent variations. HIGHLIGHTS Brain-derived tau (BD-tau) is a novel blood-based biomarker that quantifies tau protein of CNS origin. Effects of preanalytical handling procedures on the quality and reproducibility of BD-tau measures are unknown. In two cohorts of n = 105 participants, we compared BD-tau concentrations and diagnostic performances in paired plasma and serum samples, and evaluated impacts of batch-to-batch reagent variations. Paired plasma and serum showed equivalent diagnostic performances to separate amyloid-positive AD from amyloid-negative controls, indicating both can be used independently. Repeated measurements and longitudinal trajectories of plasma BD-tau were unaffected by batch-to-batch reagent variation.
Collapse
Affiliation(s)
- Fernando Gonzalez-Ortiz
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 90, Sweden
| | - Alexandre Dias
- Institute for Research & Innovation in Health (i3S), 4200-135, Porto, Portugal
| | - Michael Turton
- Bioventix Plc, 7 Romans Business Park, East Street, Farnham, Surrey GU9 7SX, UK
| | - Rui Magalhães
- Population Studies, Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Przemysław R. Kac
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 90, Sweden
| | - Manuel Correia
- Department of Neurology, Centro Hospitalar Universitário do Porto, Porto 4050-101, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Peter Harrison
- Bioventix Plc, 7 Romans Business Park, East Street, Farnham, Surrey GU9 7SX, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 90, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal 431 80, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London WC1N 3BG, United Kingdom
- UK Dementia Research Institute at UCL, London WC1N 3BG, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, 17 Science Park W Ave, Science Park, Hong Kong, China
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison 53726, WI, USA
| | - Luís F. Maia
- Institute for Research & Innovation in Health (i3S), 4200-135, Porto, Portugal
- Department of Neurology, Centro Hospitalar Universitário do Porto, Porto 4050-101, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 90, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal 431 80, Sweden
| | - Thomas K. Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 90, Sweden
- Department of Psychiatry, University of Pittsburgh, Pittsburgh 15213, PA, USA
| |
Collapse
|
29
|
Wang ZB, Tan L, Gao PY, Ma YH, Fu Y, Sun Y, Yu JT. Associations of the A/T/N profiles in PET, CSF, and plasma biomarkers with Alzheimer's disease neuropathology at autopsy. Alzheimers Dement 2023; 19:4421-4435. [PMID: 37506291 DOI: 10.1002/alz.13413] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/03/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023]
Abstract
INTRODUCTION To examine the extent to which positron emission tomography (PET)-, cerebrospinal fluid (CSF)-, and plasma-related amyloid-β/tau/neurodegeneration (A/T/N) biomarkers are associated with Alzheimer's disease (AD) neuropathology at autopsy. METHODS A total of 100 participants who respectively underwent antemortem biomarker measurements and postmortem neuropathology were included in the Alzheimer's Disease Neuroimaging Initiative (ADNI). We examined the associations of PET-, CSF-, and plasma-related A/T/N biomarkers in combinations or alone with AD neuropathological changes (ADNC). RESULTS PET- and CSF-related A/T/N biomarkers in combination showed high concordance with the ADNC stage and alone showed high accuracy in discriminating autopsy-confirmed AD. However, the plasma-related A/T/N biomarkers alone showed better discriminative performance only when combined with apolipoprotein E (APO)E ε4 genotype. DISCUSSION This study supports that PET- and CSF-related A/T/N profiles can be used to predict accurately the stages of AD neuropathology. For diagnostic settings, PET-, CSF-, and plasma-related A/T/N biomarkers are all useful diagnostic tools to detect the presence of AD neuropathology. HIGHLIGHTS PET- and CSF-related A/T/N biomarkers in combination can accurately predict the specific stages of AD neuropathology. PET- and CSF-related A/T/N biomarkers alone may serve as a precise diagnostic tool for detecting AD neuropathology at autopsy. Plasma-related A/T/N biomarkers may need combined risk factors when used as a diagnostic tool. Aβ PET and CSF p-tau181/Aβ42 were most consistent with Aβ pathology, while tau PET and CSF p-tau181/Aβ42 were most consistent with tau pathology.
Collapse
Affiliation(s)
- Zhi-Bo Wang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Pei-Yang Gao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan Sun
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Wang ZB, Tan L, Wang HF, Chen SD, Fu Y, Gao PY, Ma YH, Guo Y, Hou JH, Zhang DD, Yu JT. Differences between ante mortem Alzheimer's disease biomarkers in predicting neuropathology at autopsy. Alzheimers Dement 2023; 19:3613-3624. [PMID: 36840620 DOI: 10.1002/alz.12997] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 02/26/2023]
Abstract
INTRODUCTION This study aimed to assess whether biomarkers related to amyloid, tau, and neurodegeneration can accurately predict Alzheimer's disease (AD) neuropathology at autopsy in early and late clinical stages. METHODS We included 100 participants who had ante mortem biomarker measurements and underwent post mortem neuropathological examination. Based on ante mortem clinical diagnosis, participants were divided into non-dementia and dementia, as early or late clinical stages. RESULTS Amyloid positron emission tomography (PET) and cerebrospinal fluid (CSF) amyloid beta (Aβ)42/phosphorylated tau (p-tau)181 showed excellent performance in differentiating autopsy-confirmed AD and predicting the risk of neuropathological changes in early and late clinical stages. However, CSF Aβ42 performed better in the early clinical stage, while CSF p-tau181, CSF t-tau, and plasma p-tau181 performed better in the late clinical stage. DISCUSSION Our findings provide important clinical information that, if using PET, CSF, and plasma biomarkers to detect AD pathology, researchers must consider their differential performances at different clinical stages of AD. HIGHLIGHTS Amyloid PET and CSF Aβ42/p-tau181 were the most promising candidate biomarkers for predicting AD pathology. CSF Aβ42 can serve as a candidate predictive biomarker in the early clinical stage of AD. CSF p-tau181, CSF t-tau, and plasma p-tau181 can serve as candidate predictive biomarkers in the late clinical stage of AD. Combining APOE ε4 genotypes can significantly improve the predictive accuracy of AD-related biomarkers for AD pathology.
Collapse
Affiliation(s)
- Zhi-Bo Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Hui-Fu Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Pei-Yang Gao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia-Hui Hou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Dan-Dan Zhang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
31
|
Doher N, Davoudi V, Magaki S, Townley RA, Haeri M, Vinters HV. Illustrated Neuropathologic Diagnosis of Alzheimer's Disease. Neurol Int 2023; 15:857-867. [PMID: 37489360 PMCID: PMC10366902 DOI: 10.3390/neurolint15030054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 07/26/2023] Open
Abstract
As of 2022, the prevalence of Alzheimer's disease (AD) among individuals aged 65 and older is estimated to be 6.2 million in the United States. This figure is predicted to grow to 13.8 million by 2060. An accurate assessment of neuropathologic changes represents a critical step in understanding the underlying mechanisms in AD. The current method for assessing postmortem Alzheimer's disease neuropathologic change follows version 11 of the National Alzheimer's Coordinating Center (NACC) coding guidebook. Ambiguity regarding steps in the ABC scoring method can lead to increased time or inaccuracy in staging AD. We present a concise overview of how this postmortem diagnosis is made and relate it to the evolving understanding of antemortem AD biomarkers.
Collapse
Affiliation(s)
- Nicholas Doher
- Department of Neurology, University of Florida, Gainesville, FL 32611, USA
| | - Vahid Davoudi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Shino Magaki
- Department of Pathology and Laboratory Medicine, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA
| | - Ryan A Townley
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- The University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway City, KS 66205, USA
| | - Mohammad Haeri
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- The University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway City, KS 66205, USA
| | - Harry V Vinters
- Department of Pathology and Laboratory Medicine, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA
- Department of Neurology, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA
- Brain Research Institute, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
32
|
Wisch JK, Butt OH, Gordon BA, Schindler SE, Fagan AM, Henson RL, Yang C, Boerwinkle AH, Benzinger TLS, Holtzman DM, Morris JC, Cruchaga C, Ances BM. Proteomic clusters underlie heterogeneity in preclinical Alzheimer's disease progression. Brain 2023; 146:2944-2956. [PMID: 36542469 PMCID: PMC10316757 DOI: 10.1093/brain/awac484] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 11/21/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
Heterogeneity in progression to Alzheimer's disease (AD) poses challenges for both clinical prognosis and clinical trial implementation. Multiple AD-related subtypes have previously been identified, suggesting differences in receptivity to drug interventions. We identified early differences in preclinical AD biomarkers, assessed patterns for developing preclinical AD across the amyloid-tau-(neurodegeneration) [AT(N)] framework, and considered potential sources of difference by analysing the CSF proteome. Participants (n = 10) enrolled in longitudinal studies at the Knight Alzheimer Disease Research Center completed four or more lumbar punctures. These individuals were cognitively normal at baseline. Cerebrospinal fluid measures of amyloid-β (Aβ)42, phosphorylated tau (pTau181), and neurofilament light chain (NfL) as well as proteomics values were evaluated. Imaging biomarkers, including PET amyloid and tau, and structural MRI, were repeatedly obtained when available. Individuals were staged according to the amyloid-tau-(neurodegeneration) framework. Growth mixture modelling, an unsupervised clustering technique, identified three patterns of biomarker progression as measured by CSF pTau181 and Aβ42. Two groups (AD Biomarker Positive and Intermediate AD Biomarker) showed distinct progression from normal biomarker status to having biomarkers consistent with preclinical AD. A third group (AD Biomarker Negative) did not develop abnormal AD biomarkers over time. Participants grouped by CSF trajectories were re-classified using only proteomic profiles (AUCAD Biomarker Positive versus AD Biomarker Negative = 0.857, AUCAD Biomarker Positive versus Intermediate AD Biomarkers = 0.525, AUCIntermediate AD Biomarkers versus AD Biomarker Negative = 0.952). We highlight heterogeneity in the development of AD biomarkers in cognitively normal individuals. We identified some individuals who became amyloid positive before the age of 50 years. A second group, Intermediate AD Biomarkers, developed elevated CSF ptau181 significantly before becoming amyloid positive. A third group were AD Biomarker Negative over repeated testing. Our results could influence the selection of participants for specific treatments (e.g. amyloid-reducing versus other agents) in clinical trials. CSF proteome analysis highlighted additional non-AT(N) biomarkers for potential therapies, including blood-brain barrier-, vascular-, immune-, and neuroinflammatory-related targets.
Collapse
Affiliation(s)
- Julie K Wisch
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Omar H Butt
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Brian A Gordon
- Department of Radiology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Hope Center, Washington University in Saint Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anne M Fagan
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rachel L Henson
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Chengran Yang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anna H Boerwinkle
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Hope Center, Washington University in Saint Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John C Morris
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carlos Cruchaga
- Hope Center, Washington University in Saint Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Beau M Ances
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Hope Center, Washington University in Saint Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
33
|
Bermudez C, Graff-Radford J, Syrjanen JA, Stricker NH, Algeciras-Schimnich A, Kouri N, Kremers WK, Petersen RC, Jack CR, Knopman DS, Dickson DW, Nguyen AT, Reichard RR, Murray ME, Mielke MM, Vemuri P. Plasma biomarkers for prediction of Alzheimer's disease neuropathologic change. Acta Neuropathol 2023; 146:13-29. [PMID: 37269398 PMCID: PMC10478071 DOI: 10.1007/s00401-023-02594-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/14/2023] [Accepted: 05/24/2023] [Indexed: 06/05/2023]
Abstract
While plasma biomarkers for Alzheimer's disease (AD) are increasingly being evaluated for clinical diagnosis and prognosis, few population-based autopsy studies have evaluated their utility in the context of predicting neuropathological changes. Our goal was to investigate the utility of clinically available plasma markers in predicting Braak staging, neuritic plaque score, Thal phase, and overall AD neuropathological change (ADNC).We utilized a population-based prospective study of 350 participants with autopsy and antemortem plasma biomarker testing using clinically available antibody assay (Quanterix) consisting of Aβ42/40 ratio, p-tau181, GFAP, and NfL. We utilized a variable selection procedure in cross-validated (CV) logistic regression models to identify the best set of plasma predictors along with demographic variables, and a subset of neuropsychological tests comprising the Mayo Clinic Preclinical Alzheimer Cognitive Composite (Mayo-PACC). ADNC was best predicted with plasma GFAP, NfL, p-tau181 biomarkers along with APOE ε4 carrier status and Mayo-PACC cognitive score (CV AUC = 0.798). Braak staging was best predicted using plasma GFAP, p-tau181, and cognitive scores (CV AUC = 0.774). Neuritic plaque score was best predicted using plasma Aβ42/40 ratio, p-tau181, GFAP, and NfL biomarkers (CV AUC = 0.770). Thal phase was best predicted using GFAP, NfL, p-tau181, APOE ε4 carrier status and Mayo-PACC cognitive score (CV AUC = 0.754). We found that GFAP and p-tau provided non-overlapping information on both neuritic plaque and Braak stage scores whereas Aβ42/40 and NfL were mainly useful for prediction of neuritic plaque scores. Separating participants by cognitive status improved predictive performance, particularly when plasma biomarkers were included. Plasma biomarkers can differentially inform about overall ADNC pathology, Braak staging, and neuritic plaque score when combined with demographics and cognitive variables and have significant utility for earlier detection of AD.
Collapse
Affiliation(s)
- Camilo Bermudez
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55902, USA.
| | | | - Jeremy A Syrjanen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Nikki H Stricker
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | | | - Naomi Kouri
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Walter K Kremers
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Ronald C Petersen
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55902, USA
| | | | - David S Knopman
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55902, USA
| | | | - Aivi T Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - R Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Michelle M Mielke
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | | |
Collapse
|
34
|
Telser J, Grossmann K, Wohlwend N, Risch L, Saely CH, Werner P. Phosphorylated tau in Alzheimer's disease. Adv Clin Chem 2023; 116:31-111. [PMID: 37852722 DOI: 10.1016/bs.acc.2023.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
There is a need for blood biomarkers to detect individuals at different Alzheimer's disease (AD) stages because obtaining cerebrospinal fluid-based biomarkers is invasive and costly. Plasma phosphorylated tau proteins (p-tau) have shown potential as such biomarkers. This systematic review was conducted according to the PRISMA guidelines and aimed to determine whether quantification of plasma tau phosphorylated at threonine 181 (p-tau181), threonine 217 (p-tau217) and threonine 231 (p-tau231) is informative in the diagnosis of AD. All p-tau isoforms increase as a function of Aβ-accumulation and discriminate healthy individuals from those at preclinical AD stages with high accuracy. P-tau231 increases earliest, followed by p-tau181 and p-tau217. In advanced stages, all p-tau isoforms are associated with the clinical classification of AD and increase with disease severity, with the greatest increase seen for p-tau217. This is also reflected by a better correlation of p-tau217 with Aβ scans, whereas both, p-tau217 and p-tau181 correlated equally with tau scans. However, at the very advanced stages, p-tau181 begins to plateau, which may mirror the trajectory of the Aβ pathology and indicate an association with a more intermediate risk of AD. Across the AD continuum, the incremental increase in all biomarkers is associated with structural changes in widespread brain regions and underlying cognitive decline. Furthermore, all isoforms differentiate AD from non-AD neurodegenerative disorders, making them specific for AD. Incorporating p-tau181, p-tau217 and p-tau231 in clinical use requires further studies to examine ideal cut-points and harmonize assays.
Collapse
Affiliation(s)
- Julia Telser
- Faculty of Medical Science, Private University in the Principality of Liechtenstein, Triesen, Liechtenstein; Laboratory Dr. Risch, Vaduz, Liechtenstein
| | - Kirsten Grossmann
- Faculty of Medical Science, Private University in the Principality of Liechtenstein, Triesen, Liechtenstein; Laboratory Dr. Risch, Vaduz, Liechtenstein
| | - Niklas Wohlwend
- Laboratory Dr. Risch, Vaduz, Liechtenstein; Department of Internal Medicine Spital Grabs, Spitalregion Rheintal Werdenberg Sarganserland, Grabs, Switzerland
| | - Lorenz Risch
- Faculty of Medical Science, Private University in the Principality of Liechtenstein, Triesen, Liechtenstein; Laboratory Dr. Risch, Vaduz, Liechtenstein; University Institute of Clinical Chemistry, University Hospital and University of Bern, Inselspital, Bern, Switzerland
| | - Christoph H Saely
- Faculty of Medical Science, Private University in the Principality of Liechtenstein, Triesen, Liechtenstein; Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
| | - Philipp Werner
- Department of Neurology, State Hospital of Rankweil, Academic Teaching Hospital, Rankweil, Austria.
| |
Collapse
|
35
|
Zhang W, Young JI, Gomez L, Schmidt MA, Lukacsovich D, Varma A, Chen XS, Martin ER, Wang L. Distinct CSF biomarker-associated DNA methylation in Alzheimer's disease and cognitively normal subjects. Alzheimers Res Ther 2023; 15:78. [PMID: 37038196 PMCID: PMC10088180 DOI: 10.1186/s13195-023-01216-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 03/21/2023] [Indexed: 04/12/2023]
Abstract
BACKGROUND Growing evidence has demonstrated that DNA methylation (DNAm) plays an important role in Alzheimer's disease (AD) and that DNAm differences can be detected in the blood of AD subjects. Most studies have correlated blood DNAm with the clinical diagnosis of AD in living individuals. However, as the pathophysiological process of AD can begin many years before the onset of clinical symptoms, there is often disagreement between neuropathology in the brain and clinical phenotypes. Therefore, blood DNAm associated with AD neuropathology, rather than with clinical data, would provide more relevant information on AD pathogenesis. METHODS We performed a comprehensive analysis to identify blood DNAm associated with cerebrospinal fluid (CSF) pathological biomarkers for AD. Our study included matched samples of whole blood DNA methylation, CSF Aβ42, phosphorylated tau181 (pTau181), and total tau (tTau) biomarkers data, measured on the same subjects and at the same clinical visits from a total of 202 subjects (123 CN or cognitively normal, 79 AD) in the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort. To validate our findings, we also examined the association between premortem blood DNAm and postmortem brain neuropathology measured on a group of 69 subjects in the London dataset. RESULTS We identified a number of novel associations between blood DNAm and CSF biomarkers, demonstrating that changes in pathological processes in the CSF are reflected in the blood epigenome. Overall, the CSF biomarker-associated DNAm is relatively distinct in CN and AD subjects, highlighting the importance of analyzing omics data measured on cognitively normal subjects (which includes preclinical AD subjects) to identify diagnostic biomarkers, and considering disease stages in the development and testing of AD treatment strategies. Moreover, our analysis revealed biological processes associated with early brain impairment relevant to AD are marked by DNAm in the blood, and blood DNAm at several CpGs in the DMR on HOXA5 gene are associated with pTau181 in the CSF, as well as tau-pathology and DNAm in the brain, nominating DNAm at this locus as a promising candidate AD biomarker. CONCLUSIONS Our study provides a valuable resource for future mechanistic and biomarker studies of DNAm in AD.
Collapse
Affiliation(s)
- Wei Zhang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami Miller School of Medicine, 1120 NW 14Th Street, Miami, FL, 33136, USA
| | - Juan I Young
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Lissette Gomez
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Michael A Schmidt
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - David Lukacsovich
- Division of Biostatistics, Department of Public Health Sciences, University of Miami Miller School of Medicine, 1120 NW 14Th Street, Miami, FL, 33136, USA
| | - Achintya Varma
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - X Steven Chen
- Division of Biostatistics, Department of Public Health Sciences, University of Miami Miller School of Medicine, 1120 NW 14Th Street, Miami, FL, 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Eden R Martin
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Lily Wang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami Miller School of Medicine, 1120 NW 14Th Street, Miami, FL, 33136, USA.
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
36
|
Grothe MJ, Moscoso A, Silva-Rodríguez J, Lange C, Nho K, Saykin AJ, Nelson PT, Schöll M, Buchert R, Teipel S. Differential diagnosis of amnestic dementia patients based on an FDG-PET signature of autopsy-confirmed LATE-NC. Alzheimers Dement 2023; 19:1234-1244. [PMID: 35971593 PMCID: PMC9929029 DOI: 10.1002/alz.12763] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/13/2022] [Accepted: 07/13/2022] [Indexed: 11/07/2022]
Abstract
INTRODUCTION Limbic age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) is common in advanced age and can underlie a clinical presentation mimicking Alzheimer's disease (AD). We studied whether an autopsy-derived fluorodeoxyglucose positron emission tomography (FDG-PET) signature of LATE-NC provides clinical utility for differential diagnosis of amnestic dementia patients. METHODS Ante mortem FDG-PET patterns from autopsy-confirmed LATE-NC (N = 7) and AD (N = 23) patients were used to stratify an independent cohort of clinically diagnosed AD dementia patients (N = 242) based on individual FDG-PET profiles. RESULTS Autopsy-confirmed LATE-NC and AD groups showed markedly distinct temporo-limbic and temporo-parietal FDG-PET patterns, respectively. Clinically diagnosed AD dementia patients showing a LATE-NC-like FDG-PET pattern (N = 25, 10%) were significantly older, showed less abnormal AD biomarker levels, lower APOE ε4, and higher TMEM106B risk allele load. Clinically, they exhibited a more memory-predominant profile and a generally slower disease course. DISCUSSION An autopsy-derived temporo-limbic FDG-PET signature identifies older amnestic patients whose clinical, genetic, and molecular biomarker features are consistent with underlying LATE-NC.
Collapse
Affiliation(s)
- Michel J. Grothe
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Wallenberg Center for Molecular and Translational Medicine and Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Alexis Moscoso
- Wallenberg Center for Molecular and Translational Medicine and Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Jesús Silva-Rodríguez
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Catharina Lange
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nuclear Medicine, Berlin, Germany
| | - Kwangsik Nho
- Indiana Alzheimer’s Disease Research Center and Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Andrew J. Saykin
- Indiana Alzheimer’s Disease Research Center and Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Peter T. Nelson
- Sanders-Brown Center on Aging and Department of Pathology, University of Kentucky, Lexington, Kentucky, USA
| | - Michael Schöll
- Wallenberg Center for Molecular and Translational Medicine and Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Ralph Buchert
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | | |
Collapse
|
37
|
Gonzalez-Ortiz F, Kac PR, Brum WS, Zetterberg H, Blennow K, Karikari TK. Plasma phospho-tau in Alzheimer's disease: towards diagnostic and therapeutic trial applications. Mol Neurodegener 2023; 18:18. [PMID: 36927491 PMCID: PMC10022272 DOI: 10.1186/s13024-023-00605-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/15/2023] [Indexed: 03/18/2023] Open
Abstract
As the leading cause of dementia, Alzheimer's disease (AD) is a major burden on affected individuals, their families and caregivers, and healthcare systems. Although AD can be identified and diagnosed by cerebrospinal fluid or neuroimaging biomarkers that concord with neuropathological evidence and clinical symptoms, challenges regarding practicality and accessibility hinder their widespread availability and implementation. Consequently, many people with suspected cognitive impairment due to AD do not receive a biomarker-supported diagnosis. Blood biomarkers have the capacity to help expand access to AD diagnostics worldwide. One such promising biomarker is plasma phosphorylated tau (p-tau), which has demonstrated specificity to AD versus non-AD neurodegenerative diseases, and will be extremely important to inform on clinical diagnosis and eligibility for therapies that have recently been approved. This review provides an update on the diagnostic and prognostic performances of plasma p-tau181, p-tau217 and p-tau231, and their associations with in vivo and autopsy-verified diagnosis and pathological hallmarks. Additionally, we discuss potential applications and unanswered questions of plasma p-tau for therapeutic trials, given their recent addition to the biomarker toolbox for participant screening, recruitment and during-trial monitoring. Outstanding questions include assay standardization, threshold generation and biomarker verification in diverse cohorts reflective of the wider community attending memory clinics and included in clinical trials.
Collapse
Affiliation(s)
- Fernando Gonzalez-Ortiz
- grid.8761.80000 0000 9919 9582Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- grid.1649.a000000009445082XClinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Przemysław R. Kac
- grid.8761.80000 0000 9919 9582Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Wagner S. Brum
- grid.8761.80000 0000 9919 9582Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- grid.8532.c0000 0001 2200 7498Graduate Program in Biological Sciences: Biochemistry, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil
| | - Henrik Zetterberg
- grid.8761.80000 0000 9919 9582Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- grid.1649.a000000009445082XClinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- grid.83440.3b0000000121901201Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- grid.83440.3b0000000121901201UK Dementia Research Institute at UCL, London, UK
- grid.24515.370000 0004 1937 1450Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- grid.8761.80000 0000 9919 9582Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- grid.1649.a000000009445082XClinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Thomas K. Karikari
- grid.8761.80000 0000 9919 9582Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- grid.21925.3d0000 0004 1936 9000Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA USA
| |
Collapse
|
38
|
Salvadó G, Ossenkoppele R, Ashton NJ, Beach TG, Serrano GE, Reiman EM, Zetterberg H, Mattsson-Carlgren N, Janelidze S, Blennow K, Hansson O. Specific associations between plasma biomarkers and postmortem amyloid plaque and tau tangle loads. EMBO Mol Med 2023; 15:e17123. [PMID: 36912178 PMCID: PMC10165361 DOI: 10.15252/emmm.202217123] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/21/2023] [Accepted: 02/21/2023] [Indexed: 03/14/2023] Open
Abstract
Several promising plasma biomarkers for Alzheimer's disease have been recently developed, but their neuropathological correlates have not yet been fully determined. To investigate and compare independent associations between multiple plasma biomarkers (p-tau181, p-tau217, p-tau231, Aβ42/40, GFAP, and NfL) and neuropathologic measures of amyloid and tau, we included 105 participants from the Arizona Study of Aging and Neurodegenerative Disorders (AZSAND) with antemortem plasma samples and a postmortem neuropathological exam, 48 of whom had longitudinal p-tau217 and p-tau181. When simultaneously including plaque and tangle loads, the Aβ42/40 ratio and p-tau231 were only associated with plaques (ρAβ42/40 [95%CI] = -0.53[-0.65, -0.35], ρp-tau231 [95%CI] = 0.28[0.10, 0.43]), GFAP was only associated with tangles (ρGFAP [95%CI] = 0.39[0.17, 0.57]), and p-tau217 and p-tau181 were associated with both plaques (ρp-tau217 [95%CI] = 0.40[0.21, 0.56], ρp-tau181 [95%CI] = 0.36[0.15, 0.50]) and tangles (ρp-tau217 [95%CI] = 0.52[0.34, 0.66]; ρp-tau181 [95%CI] = 0.36[0.17, 0.52]). A model combining p-tau217 and the Aβ42/40 ratio showed the highest accuracy for predicting the presence of Alzheimer's disease neuropathological change (ADNC, AUC[95%CI] = 0.89[0.82, 0.96]) and plaque load (R2 = 0.55), while p-tau217 alone was optimal for predicting tangle load (R2 = 0.45). Our results suggest that high-performing assays of plasma p-tau217 and Aβ42/40 might be an optimal combination to assess Alzheimer's-related pathology in vivo.
Collapse
Affiliation(s)
- Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.,Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley, NHS Foundation, London, UK
| | | | | | - Eric M Reiman
- Banner Alzheimer's Institute, Arizona State University and University of Arizona, Phoenix, AZ, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK.,UK Dementia Research Institute at UCL, London, UK.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW Several plasma biomarkers for Alzheimer's disease and related disorders (ADRD) have demonstrated clinical and technical robustness. However, are they ready for clinical implementation? This review critically appraises current evidence for and against the immediate use of plasma biomarkers in clinical care. RECENT FINDINGS Plasma biomarkers have significantly improved our understanding of ADRD time-course, risk factors, diagnosis and prognosis. These advances are accelerating the development and in-human testing of therapeutic candidates, and the selection of individuals with subtle biological evidence of disease who fit the criteria for early therapeutic targeting. However, standardized tests and well validated cut-off values are lacking. Moreover, some assays (e.g., plasma Aβ methods) have poor robustness to withstand inevitable day-to-day technical variations. Additionally, recent reports suggest that common comorbidities of aging (e.g., kidney disease, diabetes, hypertension) can erroneously affect plasma biomarker levels, clinical utility and generalizability. Furthermore, it is unclear if health disparities can explain reported racial/ethnic differences in biomarker levels and functions. Finally, current clinically approved plasma methods are more expensive than CSF assays, questioning their cost effectiveness. SUMMARY Plasma biomarkers have biological and clinical capacity to detect ADRD. However, their widespread use requires issues around thresholds, comorbidities and diverse populations to be addressed.
Collapse
Affiliation(s)
- Wasiu G. Balogun
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology
- 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, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Thomas K. Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
40
|
Cullen NC, Janelidze S, Mattsson-Carlgren N, Palmqvist S, Bittner T, Suridjan I, Jethwa A, Kollmorgen G, Brum WS, Zetterberg H, Blennow K, Stomrud E, Hansson O. Test-retest variability of plasma biomarkers in Alzheimer's disease and its effects on clinical prediction models. Alzheimers Dement 2023; 19:797-806. [PMID: 35699240 PMCID: PMC9747985 DOI: 10.1002/alz.12706] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/22/2022] [Accepted: 04/27/2022] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The effect of random error on the performance of blood-based biomarkers for Alzheimer's disease (AD) must be determined before clinical implementation. METHODS We measured test-retest variability of plasma amyloid beta (Aβ)42/Aβ40, neurofilament light (NfL), glial fibrillary acidic protein (GFAP), and phosphorylated tau (p-tau)217 and simulated effects of this variability on biomarker performance when predicting either cerebrospinal fluid (CSF) Aβ status or conversion to AD dementia in 399 non-demented participants with cognitive symptoms. RESULTS Clinical performance was highest when combining all biomarkers. Among single-biomarkers, p-tau217 performed best. Test-retest variability ranged from 4.1% (Aβ42/Aβ40) to 25% (GFAP). This variability reduced the performance of the biomarkers (≈ΔAUC [area under the curve] -1% to -4%) with the least effects on models with p-tau217. The percent of individuals with unstable predicted outcomes was lowest for the multi-biomarker combination (14%). DISCUSSION Clinical prediction models combining plasma biomarkers-particularly p-tau217-exhibit high performance and are less effected by random error. Individuals with unstable predicted outcomes ("gray zone") should be recommended for further tests.
Collapse
Affiliation(s)
- Nicholas C. Cullen
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | | | | | | | | | - Wagner S. Brum
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, U.K
- UK Dementia Research Institute at UCL, London, United Kingdom
- Hong Kong Center for 27 Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
41
|
Gonzalez-Ortiz F, Turton M, Kac PR, Smirnov D, Premi E, Ghidoni R, Benussi L, Cantoni V, Saraceno C, Rivolta J, Ashton NJ, Borroni B, Galasko D, Harrison P, Zetterberg H, Blennow K, Karikari TK. Brain-derived tau: a novel blood-based biomarker for Alzheimer's disease-type neurodegeneration. Brain 2023; 146:1152-1165. [PMID: 36572122 PMCID: PMC9976981 DOI: 10.1093/brain/awac407] [Citation(s) in RCA: 108] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 09/23/2022] [Accepted: 09/25/2022] [Indexed: 12/28/2022] Open
Abstract
Blood-based biomarkers for amyloid beta and phosphorylated tau show good diagnostic accuracies and agreements with their corresponding CSF and neuroimaging biomarkers in the amyloid/tau/neurodegeneration [A/T/(N)] framework for Alzheimer's disease. However, the blood-based neurodegeneration marker neurofilament light is not specific to Alzheimer's disease while total-tau shows lack of correlation with CSF total-tau. Recent studies suggest that blood total-tau originates principally from peripheral, non-brain sources. We sought to address this challenge by generating an anti-tau antibody that selectively binds brain-derived tau and avoids the peripherally expressed 'big tau' isoform. We applied this antibody to develop an ultrasensitive blood-based assay for brain-derived tau, and validated it in five independent cohorts (n = 609) including a blood-to-autopsy cohort, CSF biomarker-classified cohorts and memory clinic cohorts. In paired samples, serum and CSF brain-derived tau were significantly correlated (rho = 0.85, P < 0.0001), while serum and CSF total-tau were not (rho = 0.23, P = 0.3364). Blood-based brain-derived tau showed equivalent diagnostic performance as CSF total-tau and CSF brain-derived tau to separate biomarker-positive Alzheimer's disease participants from biomarker-negative controls. Furthermore, plasma brain-derived tau accurately distinguished autopsy-confirmed Alzheimer's disease from other neurodegenerative diseases (area under the curve = 86.4%) while neurofilament light did not (area under the curve = 54.3%). These performances were independent of the presence of concomitant pathologies. Plasma brain-derived tau (rho = 0.52-0.67, P = 0.003), but not neurofilament light (rho = -0.14-0.17, P = 0.501), was associated with global and regional amyloid plaque and neurofibrillary tangle counts. These results were further verified in two memory clinic cohorts where serum brain-derived tau differentiated Alzheimer's disease from a range of other neurodegenerative disorders, including frontotemporal lobar degeneration and atypical parkinsonian disorders (area under the curve up to 99.6%). Notably, plasma/serum brain-derived tau correlated with neurofilament light only in Alzheimer's disease but not in the other neurodegenerative diseases. Across cohorts, plasma/serum brain-derived tau was associated with CSF and plasma AT(N) biomarkers and cognitive function. Brain-derived tau is a new blood-based biomarker that outperforms plasma total-tau and, unlike neurofilament light, shows specificity to Alzheimer's disease-type neurodegeneration. Thus, brain-derived tau demonstrates potential to complete the AT(N) scheme in blood, and will be useful to evaluate Alzheimer's disease-dependent neurodegenerative processes for clinical and research purposes.
Collapse
Affiliation(s)
- Fernando Gonzalez-Ortiz
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Michael Turton
- Bioventix Plc, Romans Business Park, Farnham, Surrey GU9 7SX, UK
| | - Przemysław R Kac
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Denis Smirnov
- University of California, San Diego and Shiely-Marcos Alzheimer’s Disease Research Center, La Jolla, CA 92037, USA
| | - Enrico Premi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, BS 25121, Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia 25121, Italy
| | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia 25121, Italy
| | - Valentina Cantoni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, BS 25121, Italy
| | - Claudia Saraceno
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia 25121, Italy
| | - Jasmine Rivolta
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, BS 25121, Italy
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg 405 30, Sweden
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, SE5 8AF, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, SE5 8AF, UK
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, BS 25121, Italy
| | - Douglas Galasko
- University of California, San Diego and Shiely-Marcos Alzheimer’s Disease Research Center, La Jolla, CA 92037, USA
| | - Peter Harrison
- Bioventix Plc, Romans Business Park, Farnham, Surrey GU9 7SX, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 431 80, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Shatin, N.T., Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 431 80, Sweden
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
42
|
Gorelick PB. Blood and Cerebrospinal Fluid Biomarkers in Vascular Dementia and Alzheimer's Disease: A Brief Review. Clin Geriatr Med 2023; 39:67-76. [PMID: 36404033 DOI: 10.1016/j.cger.2022.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The Maintenance of brain health is a lifelong process whereby potentially deleterious exposures such as cardiovascular risks, amyloid beta, and phosphorylated tau may adversely affect the brain decades before there are clinical manifestations. Thus, the early structural and neuropathological foundation for the development of cognitive impairment and its allied features later in life may provide precursor targets such that interventions may be applied to prevent or slow cognitively impairing processes if the underlying mechanism(s) can be addressed in time.
Collapse
Affiliation(s)
- Philip B Gorelick
- Section of Stroke and Neurocritical Care, Davee Department of Neurology, Northwestern University Feinberg School of Medicine, 625 North Michigan Avenue Suite 1150, Chicago, IL 60611, USA.
| |
Collapse
|
43
|
Leuzy A, Mattsson-Carlgren N, Cullen NC, Stomrud E, Palmqvist S, La Joie R, Iaccarino L, Zetterberg H, Rabinovici G, Blennow K, Janelidze S, Hansson O. Robustness of CSF Aβ42/40 and Aβ42/P-tau181 measured using fully automated immunoassays to detect AD-related outcomes. Alzheimers Dement 2023. [PMID: 36681387 DOI: 10.1002/alz.12897] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/26/2022] [Accepted: 11/10/2022] [Indexed: 01/23/2023]
Abstract
INTRODUCTION This study investigated the comparability of cerebrospinal fluid (CSF) cutoffs for Elecsys immunoassays for amyloid beta (Aβ)42/Aβ40 or Aβ42/phosphorylated tau (p-tau)181 and the effects of measurement variability when predicting Alzheimer's disease (AD)-related outcomes (i.e., Aβ-positron emission tomography [PET] visual read and AD neuropathology). METHODS We studied 750 participants (BioFINDER study, Alzheimer's Disease Neuroimaging Initiative [ADNI], and University of California San Francisco [UCSF]). Youden's index was used to identify cutoffs and to calculate accuracy (Aβ-PET visual read as outcome). Using longitudinal variability in Aβ-negative controls, we identified a gray zone around cut-points where the risk of an inconsistent predicted outcome was >5%. RESULTS For Aβ42/Aβ40, cutoffs across cohorts were <0.059 (BioFINDER), <0.057 (ADNI), and <0.058 (UCSF). For Aβ42/p-tau181, cutoffs were <41.90 (BioFINDER), <39.20 (ADNI), and <46.02 (UCSF). Accuracy was ≈90% for both Aβ42/Aβ40 and Aβ42/p-tau181 using these cutoffs. Using Aβ-PET as an outcome, 8.7% of participants fell within a gray zone interval for Aβ42/Aβ40, compared to 4.5% for Aβ42/p-tau181. Similar findings were observed using a measure of overall AD neuropathologic change (7.7% vs. 3.3%). In a subset with CSF and plasma Aβ42/40, the number of individuals within the gray zone was ≈1.5 to 3 times greater when using plasma Aβ42/40. DISCUSSION CSF Aβ42/p-tau181 was more robust to the effects of measurement variability, suggesting that it may be the preferred Elecsys-based measure in clinical practice and trials.
Collapse
Affiliation(s)
- Antoine Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Nicholas C Cullen
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Leonardo Iaccarino
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute at UCL, London, UK.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Gil Rabinovici
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA.,Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, USA.,Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
44
|
Abildgaard A, Parkner T, Knudsen CS, Gottrup H, Klit H. Diagnostic Cut-offs for CSF β-amyloid and tau proteins in a Danish dementia clinic. Clin Chim Acta 2023; 539:244-249. [PMID: 36572135 DOI: 10.1016/j.cca.2022.12.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND Analysis of beta-amyloid 1-42 (Aβ42), total tau (t-tau) and phosphorylated-tau 181 (p-tau) in the cerebrospinal fluid (CSF) is often performed as a part of the diagnostic work-up in case of suspected Alzheimer's dementia (AD). Unfortunately, studies on optimal CSF biomarker cut-offs in a real-world clinical setting are scarce. METHODS We retrospectively evaluated the biomarker levels of 264 consecutive patients referred to our dementia clinic. The biomarkers were analysed with the Elecsys(R) assays. Diagnoses were based on all available clinical information, including FDG-PET scans. RESULTS In total, we identified 233 patients diagnosed with dementia. The median MMSE score was 22 (IQR 18-25). AD pathophysiology was suspected in 156 patients, and the corresponding cut-offs based on the Youden index were: Aβ42: 903 ng/L (ROC-AUC 0.78); t-tau: 272 ng/L (ROC-AUC 0.78); p-tau: 24 ng/L (ROC-AUC 0.85); t-tau/Aβ42 ratio: 0.34 (ROC-AUC 0.91); p-tau/Aβ42 ratio: 0.029 (ROC-AUC 0.92). CONCLUSIONS We found the tau/Aβ42 ratios to possess the best diagnostic performance, but our estimated cut-off values for the ratios were somewhat higher than previously reported. Consequently, if the CSF analyses are used to support a diagnosis of AD in a heterogeneous high-prevalence cohort, adjustment of the cut-offs may be warranted.
Collapse
Affiliation(s)
- Anders Abildgaard
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, Denmark.
| | - Tina Parkner
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, Denmark
| | - Cindy Soendersoe Knudsen
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, Denmark
| | - Hanne Gottrup
- Department of Neurology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, Denmark
| | - Henriette Klit
- Department of Neurology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, Denmark
| |
Collapse
|
45
|
Murray ME, Moloney CM, Kouri N, Syrjanen JA, Matchett BJ, Rothberg DM, Tranovich JF, Sirmans TNH, Wiste HJ, Boon BDC, Nguyen AT, Reichard RR, Dickson DW, Lowe VJ, Dage JL, Petersen RC, Jack CR, Knopman DS, Vemuri P, Graff-Radford J, Mielke MM. Global neuropathologic severity of Alzheimer's disease and locus coeruleus vulnerability influences plasma phosphorylated tau levels. Mol Neurodegener 2022; 17:85. [PMID: 36575455 PMCID: PMC9795667 DOI: 10.1186/s13024-022-00578-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 10/26/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Advances in ultrasensitive detection of phosphorylated tau (p-tau) in plasma has enabled the use of blood tests to measure Alzheimer's disease (AD) biomarker changes. Examination of postmortem brains of participants with antemortem plasma p-tau levels remains critical to understanding comorbid and AD-specific contribution to these biomarker changes. METHODS We analyzed 35 population-based Mayo Clinic Study of Aging participants with plasma p-tau at threonine 181 and threonine 217 (p-tau181, p-tau217) available within 3 years of death. Autopsied participants included cognitively unimpaired, mild cognitive impairment, AD dementia, and non-AD neurodegenerative disorders. Global neuropathologic scales of tau, amyloid-β, TDP-43, and cerebrovascular disease were examined. Regional digital pathology measures of tau (phosphorylated threonine 181 and 217 [pT181, pT217]) and amyloid-β (6F/3D) were quantified in hippocampus and parietal cortex. Neurotransmitter hubs reported to influence development of tangles (nucleus basalis of Meynert) and amyloid-β plaques (locus coeruleus) were evaluated. RESULTS The strongest regional associations were with parietal cortex for tau burden (p-tau181 R = 0.55, p = 0.003; p-tau217 R = 0.66, p < 0.001) and amyloid-β burden (p-tau181 R = 0.59, p < 0.001; p-tau217 R = 0.71, p < 0.001). Linear regression analysis of global neuropathologic scales explained 31% of variability in plasma p-tau181 (Adj. R2 = 0.31) and 59% in plasma p-tau217 (Adj. R2 = 0.59). Neither TDP-43 nor cerebrovascular disease global scales independently contributed to variability. Global scales of tau pathology (β-coefficient = 0.060, p = 0.016) and amyloid-β pathology (β-coefficient = 0.080, p < 0.001) independently predicted plasma p-tau217 when modeled together with co-pathologies, but only amyloid-β (β-coefficient = 0.33, p = 0.021) significantly predicted plasma p-tau181. While nucleus basalis of Meynert neuron count/mm2 was not associated with plasma p-tau levels, a lower locus coeruleus neuron count/mm2 was associated with higher plasma p-tau181 (R = -0.50, p = 0.007) and higher plasma p-tau217 (R = -0.55, p = 0.002). Cognitive scores (Adj. R2 = 0.25-0.32) were predicted by the global tau scale, but not by the global amyloid-β scale or plasma p-tau when modeled simultaneously. CONCLUSIONS Higher soluble plasma p-tau levels may be the result of an intersection between insoluble deposits of amyloid-β and tau accumulation in brain, and may be associated with locus coeruleus degeneration.
Collapse
Affiliation(s)
- Melissa E. Murray
- Department of Neuroscience, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Christina M. Moloney
- Department of Neuroscience, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Naomi Kouri
- Department of Neuroscience, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Jeremy A. Syrjanen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN USA
| | - Billie J. Matchett
- Department of Neuroscience, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Darren M. Rothberg
- Department of Neuroscience, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Jessica F. Tranovich
- Department of Neuroscience, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Tiffany N. Hicks Sirmans
- Department of Neuroscience, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Heather J. Wiste
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN USA
| | - Baayla D. C. Boon
- Department of Neuroscience, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Aivi T. Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
| | - R. Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
| | - Dennis W. Dickson
- Department of Neuroscience, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Val J. Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN USA
| | - Jeffrey L. Dage
- Department of Neurology, Indiana University, Indianapolis, IN USA
| | | | | | | | | | | | - Michelle M. Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN USA
- Wake Forest University School of Medicine, Winston-Salem, NC USA
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest University School of Medicine, 525 Vine, 5th floor, Winston-Salem, NC 27157 USA
| |
Collapse
|
46
|
Long JM, Coble DW, Xiong C, Schindler SE, Perrin RJ, Gordon BA, Benzinger TLS, Grant E, Fagan AM, Harari O, Cruchaga C, Holtzman DM, Morris JC. Preclinical Alzheimer's disease biomarkers accurately predict cognitive and neuropathological outcomes. Brain 2022; 145:4506-4518. [PMID: 35867858 PMCID: PMC10200309 DOI: 10.1093/brain/awac250] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/30/2022] [Accepted: 07/20/2022] [Indexed: 01/24/2023] Open
Abstract
Alzheimer's disease biomarkers are widely accepted as surrogate markers of underlying neuropathological changes. However, few studies have evaluated whether preclinical Alzheimer's disease biomarkers predict Alzheimer's neuropathology at autopsy. We sought to determine whether amyloid PET imaging or CSF biomarkers accurately predict cognitive outcomes and Alzheimer's disease neuropathological findings. This study included 720 participants, 42-91 years of age, who were enrolled in longitudinal studies of memory and aging in the Washington University Knight Alzheimer Disease Research Center and were cognitively normal at baseline, underwent amyloid PET imaging and/or CSF collection within 1 year of baseline clinical assessment, and had subsequent clinical follow-up. Cognitive status was assessed longitudinally by Clinical Dementia Rating®. Biomarker status was assessed using predefined cut-offs for amyloid PET imaging or CSF p-tau181/amyloid-β42. Subsequently, 57 participants died and underwent neuropathologic examination. Alzheimer's disease neuropathological changes were assessed using standard criteria. We assessed the predictive value of Alzheimer's disease biomarker status on progression to cognitive impairment and for presence of Alzheimer's disease neuropathological changes. Among cognitively normal participants with positive biomarkers, 34.4% developed cognitive impairment (Clinical Dementia Rating > 0) as compared to 8.4% of those with negative biomarkers. Cox proportional hazards modelling indicated that preclinical Alzheimer's disease biomarker status, APOE ɛ4 carrier status, polygenic risk score and centred age influenced risk of developing cognitive impairment. Among autopsied participants, 90.9% of biomarker-positive participants and 8.6% of biomarker-negative participants had Alzheimer's disease neuropathological changes. Sensitivity was 87.0%, specificity 94.1%, positive predictive value 90.9% and negative predictive value 91.4% for detection of Alzheimer's disease neuropathological changes by preclinical biomarkers. Single CSF and amyloid PET baseline biomarkers were also predictive of Alzheimer's disease neuropathological changes, as well as Thal phase and Braak stage of pathology at autopsy. Biomarker-negative participants who developed cognitive impairment were more likely to exhibit non-Alzheimer's disease pathology at autopsy. The detection of preclinical Alzheimer's disease biomarkers is strongly predictive of future cognitive impairment and accurately predicts presence of Alzheimer's disease neuropathology at autopsy.
Collapse
Affiliation(s)
- Justin M Long
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Dean W Coble
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Division of Biostatistics, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Chengjie Xiong
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Division of Biostatistics, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Suzanne E Schindler
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Richard J Perrin
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Brian A Gordon
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Tammie L S Benzinger
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Elizabeth Grant
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Division of Biostatistics, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Anne M Fagan
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Oscar Harari
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Carlos Cruchaga
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - David M Holtzman
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - John C Morris
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| |
Collapse
|
47
|
Karikari TK. Blood Tests for Alzheimer's Disease: Increasing Efforts to Expand and Diversify Research Participation Is Critical for Widespread Validation and Acceptance. J Alzheimers Dis 2022; 90:967-974. [PMID: 35491788 PMCID: PMC9741736 DOI: 10.3233/jad-215730] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The recent academic and commercial development, and regulatory approvals, of blood-based Alzheimer's disease (AD) biomarkers are breakthrough developments of immense potential. However, clinical validation studies and therapeutic trial applications are limited almost exclusively to non-Hispanic White cohorts often including highly-educated, high-earning participants. This commentary argues that the true benefits of blood tests for AD will be realized by active inclusion of diverse groups including minoritized populations, people of socioeconomic status different from those included in existing cohorts, and residents of low- and middle-income countries. The article discusses key factors that are critical for a successful implementation of diversity programs.
Collapse
Affiliation(s)
- Thomas K. Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA,Correspondence to: Thomas K. Karikari, PhD, Assistant Professor, Clinical Neurochemistry Lab., Sahlgrenska University Hospital, House V3/SU, 43 180, Mölndal, Sweden. E-mails: ;
| |
Collapse
|
48
|
Pichet Binette A, Franzmeier N, Spotorno N, Ewers M, Brendel M, Biel D, Strandberg O, Janelidze S, Palmqvist S, Mattsson-Carlgren N, Smith R, Stomrud E, Ossenkoppele R, Hansson O. Amyloid-associated increases in soluble tau relate to tau aggregation rates and cognitive decline in early Alzheimer's disease. Nat Commun 2022; 13:6635. [PMID: 36333294 PMCID: PMC9636262 DOI: 10.1038/s41467-022-34129-4] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
For optimal design of anti-amyloid-β (Aβ) and anti-tau clinical trials, we need to better understand the pathophysiological cascade of Aβ- and tau-related processes. Therefore, we set out to investigate how Aβ and soluble phosphorylated tau (p-tau) relate to the accumulation of tau aggregates assessed with PET and subsequent cognitive decline across the Alzheimer's disease (AD) continuum. Using human cross-sectional and longitudinal neuroimaging and cognitive assessment data, we show that in early stages of AD, increased concentration of soluble CSF p-tau is strongly associated with accumulation of insoluble tau aggregates across the brain, and CSF p-tau levels mediate the effect of Aβ on tau aggregation. Further, higher soluble p-tau concentrations are mainly related to faster accumulation of tau aggregates in the regions with strong functional connectivity to individual tau epicenters. In this early stage, higher soluble p-tau concentrations is associated with cognitive decline, which is mediated by faster increase of tau aggregates. In contrast, in AD dementia, when Aβ fibrils and soluble p-tau levels have plateaued, cognitive decline is related to the accumulation rate of insoluble tau aggregates. Our data suggest that therapeutic approaches reducing soluble p-tau levels might be most favorable in early AD, before widespread insoluble tau aggregates.
Collapse
Affiliation(s)
- Alexa Pichet Binette
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden.
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Nicola Spotorno
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
| | - Michael Ewers
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Davina Biel
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Olof Strandberg
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
- Department of Neurology, Skåne University Hospital, Lund, 205 02, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
- Department of Neurology, Skåne University Hospital, Lund, 205 02, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Oskar Hansson
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
49
|
Graff-Radford J, Mielke MM, Hofrenning EI, Kouri N, Lesnick TG, Moloney CM, Rabinstein A, Cabrera-Rodriguez JN, Rothberg DM, Przybelski SA, Petersen RC, Knopman DS, Dickson DW, Jack CR, Algeciras-Schimnich A, Nguyen AT, Murray ME, Vemuri P. Association of plasma biomarkers of amyloid and neurodegeneration with cerebrovascular disease and Alzheimer's disease. Neurobiol Aging 2022; 119:1-7. [PMID: 35952440 PMCID: PMC9732897 DOI: 10.1016/j.neurobiolaging.2022.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/30/2022] [Accepted: 07/20/2022] [Indexed: 12/13/2022]
Abstract
The objective of this study was to determine the differential mapping of plasma biomarkers to postmortem neuropathology measures. We identified 64 participants in a population-based study with antemortem plasma markers (amyloid-β [Aβ] x-42, Aβx-40, neurofilament light [NfL], and total tau [T-tau]) who also had neuropathologic assessments of Alzheimer's and cerebrovascular pathology. We conducted weighted linear-regression models to evaluate relationships between plasma measures and neuropathology. Higher plasma NfL and Aβ42/40 ratio were associated with cerebrovascular neuropathologic scales (p < 0.05) but not with Braak stage, neuritic plaque score, or Thal phase. Plasma Aβ42/40 and NfL explained up to 18% of the variability in cerebrovascular neuropathologic scales. In participants predominantly with modest levels of Alzheimer's pathologic change, biomarkers of amyloid and neurodegeneration were associated with cerebrovascular neuropathology. NfL is a non-specific marker of brain injury, therefore its association with cerebrovascular neuropathology was expected. The association between elevated Aβ42/40 and cerebrovascular disease pathology needs further investigation but could be due to the use of less specific amyloid-β assays (x-40, x-42).
Collapse
Affiliation(s)
| | - Michelle M Mielke
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA; Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | | | - Naomi Kouri
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Timothy G Lesnick
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | - Aivi T Nguyen
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | |
Collapse
|
50
|
Morrison MS, Aparicio HJ, Blennow K, Zetterberg H, Ashton NJ, Karikari TK, Tripodis Y, Martin B, Palmisano JN, Sugarman MA, Frank B, Steinberg EG, Turk KW, Budson AE, Au R, Goldstein LE, Jun GR, Kowall NW, Killiany R, Qiu WQ, Stern RA, Mez J, McKee AC, Stein TD, Alosco ML. Ante-mortem plasma phosphorylated tau (181) predicts Alzheimer's disease neuropathology and regional tau at autopsy. Brain 2022; 145:3546-3557. [PMID: 35554506 PMCID: PMC10233293 DOI: 10.1093/brain/awac175] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/07/2022] [Accepted: 05/02/2022] [Indexed: 11/14/2022] Open
Abstract
Blood-based biomarkers such as tau phosphorylated at threonine 181 (phosphorylated-tau181) represent an accessible, cost-effective and scalable approach for the in vivo detection of Alzheimer's disease pathophysiology. Plasma-pathological correlation studies are needed to validate plasma phosphorylated-tau181 as an accurate and reliable biomarker of Alzheimer's disease neuropathological changes. This plasma-to-autopsy correlation study included participants from the Boston University Alzheimer's Disease Research Center who had a plasma sample analysed for phosphorylated-tau181 between 2008 and 2018 and donated their brain for neuropathological examination. Plasma phosphorelated-tau181 was measured with single molecule array technology. Of 103 participants, 62 (60.2%) had autopsy-confirmed Alzheimer's disease. Average time between blood draw and death was 5.6 years (standard deviation = 3.1 years). Multivariable analyses showed higher plasma phosphorylated-tau181 concentrations were associated with increased odds for having autopsy-confirmed Alzheimer's disease [AUC = 0.82, OR = 1.07, 95% CI = 1.03-1.11, P < 0.01; phosphorylated-tau standardized (z-transformed): OR = 2.98, 95% CI = 1.50-5.93, P < 0.01]. Higher plasma phosphorylated-tau181 levels were associated with increased odds for having a higher Braak stage (OR = 1.06, 95% CI = 1.02-1.09, P < 0.01) and more severe phosphorylated-tau across six cortical and subcortical brain regions (ORs = 1.03-1.06, P < 0.05). The association between plasma phosphorylated-tau181 and Alzheimer's disease was strongest in those who were demented at time of blood draw (OR = 1.25, 95%CI = 1.02-1.53), but an effect existed among the non-demented (OR = 1.05, 95% CI = 1.01-1.10). There was higher discrimination accuracy for Alzheimer's disease when blood draw occurred in years closer to death; however, higher plasma phosphorylated-tau181 levels were associated with Alzheimer's disease even when blood draw occurred >5 years from death. Ante-mortem plasma phosphorylated-tau181 concentrations were associated with Alzheimer's disease neuropathology and accurately differentiated brain donors with and without autopsy-confirmed Alzheimer's disease. These findings support plasma phosphorylated-tau181 as a scalable biomarker for the detection of Alzheimer's disease.
Collapse
Affiliation(s)
- Madeline S Morrison
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Hugo J Aparicio
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- Framingham Heart Study, Boston University School of Medicine, Boston, MA 02118, USA
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London WC1N 3BG, UK
| | - Nicholas J Ashton
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden
| | - Thomas K Karikari
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden
| | - Yorghos Tripodis
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Brett Martin
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA 02118, USA
| | - Joseph N Palmisano
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA 02118, USA
| | - Michael A Sugarman
- Department of Neurology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Brandon Frank
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Eric G Steinberg
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Katherine W Turk
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Jamaica Plain, MA 02130, USA
| | - Andrew E Budson
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Jamaica Plain, MA 02130, USA
| | - Rhoda Au
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- Framingham Heart Study, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA 02118, USA
| | - Lee E Goldstein
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Biomedical Engineering, Boston University College of Engineering, Boston, MA 02215, USA
- Department of Electrical and Computer Engineering, Boston University College of Engineering, Boston, MA 02215, USA
| | - Gyungah R Jun
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Neil W Kowall
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Jamaica Plain, MA 02130, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Ronald Killiany
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
- Center for Biomedical Imaging, Boston University School of Medicine, Boston, MA 02118, USA
| | - Wei Qiao Qiu
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Robert A Stern
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurosurgery, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jesse Mez
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- Framingham Heart Study, Boston University School of Medicine, Boston, MA 02118, USA
| | - Ann C McKee
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Jamaica Plain, MA 02130, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- VA Bedford Healthcare System, U.S. Department of Veteran Affairs, Bedford, MA 01730, USA
| | - Thor D Stein
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Jamaica Plain, MA 02130, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- VA Bedford Healthcare System, U.S. Department of Veteran Affairs, Bedford, MA 01730, USA
| | - Michael L Alosco
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|