1
|
Zhao L, Zhang M, Li Q, Wang X, Lu J, Han Y, Cai Y. Storage time affects the level and diagnostic efficacy of plasma biomarkers for neurodegenerative diseases. Neural Regen Res 2025; 20:2373-2381. [PMID: 39359094 PMCID: PMC11759026 DOI: 10.4103/nrr.nrr-d-23-01983] [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: 12/04/2023] [Revised: 02/06/2024] [Accepted: 03/15/2024] [Indexed: 10/04/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202508000-00027/figure1/v/2024-09-30T120553Z/r/image-tiff Several promising plasma biomarker proteins, such as amyloid-β (Aβ), tau, neurofilament light chain, and glial fibrillary acidic protein, are widely used for the diagnosis of neurodegenerative diseases. However, little is known about the long-term stability of these biomarker proteins in plasma samples stored at -80°C. We aimed to explore how storage time would affect the diagnostic accuracy of these biomarkers using a large cohort. Plasma samples from 229 cognitively unimpaired individuals, encompassing healthy controls and those experiencing subjective cognitive decline, as well as 99 patients with cognitive impairment, comprising those with mild cognitive impairment and dementia, were acquired from the Sino Longitudinal Study on Cognitive Decline project. These samples were stored at -80°C for up to 6 years before being used in this study. Our results showed that plasma levels of Aβ42, Aβ40, neurofilament light chain, and glial fibrillary acidic protein were not significantly correlated with sample storage time. However, the level of total tau showed a negative correlation with sample storage time. Notably, in individuals without cognitive impairment, plasma levels of total protein and tau phosphorylated protein threonine 181 (p-tau181)also showed a negative correlation with sample storage time. This was not observed in individuals with cognitive impairment. Consequently, we speculate that the diagnostic accuracy of plasma p-tau181 and the p-tau181 to total tau ratio may be influenced by sample storage time. Therefore, caution is advised when using these plasma biomarkers for the identification of neurodegenerative diseases, such as Alzheimer's disease. Furthermore, in cohort studies, it is important to consider the impact of storage time on the overall results.
Collapse
Affiliation(s)
- Lifang Zhao
- Department of Clinical Biobank, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center, Beijing, China
| | - Mingkai Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qimeng Li
- Department of Clinical Biobank, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center, Beijing, China
| | - Xuemin Wang
- Department of Clinical Biobank, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center, Beijing, China
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative diseases, Ministry of Education, Beijing, China
| | - Ying Han
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- School of Biomedical Engineering, Hainan University, Haikou, Hainan Province, China
- Center of Alzheimer’s Disease, Beijing Institute for Brain Disorders, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Beijing, China
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Gaoke Innovation Center, Shenzhen, Guangdong Province, China
| | - Yanning Cai
- Department of Clinical Biobank, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center, Beijing, China
- Key Laboratory of Neurodegenerative diseases, Ministry of Education, Beijing, China
- Department of Neurobiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Jauregi-Zinkunegi A, Wilson RE, E Langhough R, Ashton NJ, Blennow K, Johnson SC, Zetterberg H, Bruno D, Mueller KD. Associations between the logical memory test story recall metrics and plasma biomarkers for Alzheimer's disease in individuals free of dementia. Clin Neuropsychol 2025:1-20. [PMID: 40114424 DOI: 10.1080/13854046.2025.2481119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
Objective: Blood-based biomarkers are valued for their lower cost and less invasive nature, though issues with widespread implementation and accessibility remain. Process-based scores from story recall have been shown to detect neuronal network disturbances typical of Alzheimer's disease (AD) pathology more effectively than traditional metrics. This study examined the associations between process-based scores and concurrent plasma AD biomarkers in older adults without dementia, while also comparing them to traditional metrics. Additionally, it also investigated the diagnostic utility of these metrics in detecting plasma p-tau217 positivity. Methods: Data from 416 participants (mean age = 66.6 ± 7) free of dementia were extracted from the Wisconsin Registry for Alzheimer's Prevention (WRAP). Logical Memory Test (LMT) and plasma p-tau217, p-tau181, p-tau231, Aβ42/Aβ40 ratio, GFAP and NfL levels were analyzed. Bayesian regression models assessed associations between plasma biomarkers and both process-based and traditional LMT scores, controlling for the covariates. Results: The best-fitting model for plasma p-tau217 included Total ratio (Tr) and Immediate recall (BF10=573), but Tr showed stronger evidence of association (mean coefficient = 0.208; BFinclusion=14.4) than Immediate recall (mean coefficient=-0.007; BFinclusion=1.7). Tr was also the best predictor of plasma p-tau181 (mean coefficient = 0.144; BF10=10.5) and GFAP (mean coefficient = 0.141; BF10=5.8), outperforming traditional LMT scores. No memory scores were associated with plasma p-tau231 or Aβ42/40 ratio levels. Tr score was the strongest single predictor of p-tau217 positivity (BF10=38). Conclusions: These findings suggest that process-based memory scores might be useful in enhancing the detection of neuronal network disturbances associated with AD pathology, especially in settings where biomarker testing is unavailable.
Collapse
Affiliation(s)
| | - Rachael E Wilson
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
| | - Rebecca E Langhough
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Geriatric Research Education and Clinical Center of the Wm. S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Henrik Zetterberg
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, 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
| | - Davide Bruno
- School of Psychology, Liverpool John Moores University, Liverpool, UK
| | - Kimberly D Mueller
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
- Department of Communication Sciences and Disorders, University of Wisconsin - Madison, Madison, WI, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
| |
Collapse
|
3
|
Wei M, Yu X, Hu S, Hu W, Shi R, Wang M, Zhong J, Zhang Q, Zhang Y, Li C, Song Z, Jiang J, Han Y. Differences of longitudinal plasma biomarkers between single memory domain and multidomain subject cognitive decline: Evidence from SILCODE. J Alzheimers Dis 2025; 103:1060-1074. [PMID: 39791252 DOI: 10.1177/13872877241309105] [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: 01/12/2025]
Abstract
BACKGROUND Plasma biomarkers demonstrated potential in identifying amyloid pathology in early Alzheimer's disease. Different subtypes of subjective cognitive decline (SCD) may lead to different cognitive impairment conversion risks. OBJECTIVE To investigate the differences of plasma biomarkers in SCD subtypes individuals, which were unclear. METHODS The 347 individuals were involved, including 93 normal controls (NC), 76 single memory domain SCD (sd-SCD), 79 multidomain SCD (md-SCD), 55 mild cognitive impairment and 44 dementia. We investigated plasma biomarkers (Aβ42/40, p-tau181, p-tau217, NfL, and GFAP) and neuropsychological scales in the baseline and follow-up. The Kaplan-Meier survival analysis and Cox proportional hazards model were performed to investigate the risk of cognitive decline conversion. The t-test, Mann-Whitney U and multiple linear regression analysis were employed to evaluate the rate of change and correlation between PET-SUVR and plasma biomarker change. RESULTS In cognitively normal subjects, md-SCD exhibited lower Aβ42/40 and higher p-tau181 and p-tau217 levels. Kaplan-Meier survival analysis revealed that md-SCD group exhibited a higher risk of cognitive decline conversion compared to NC and sd-SCD. Within SCD subgroups, those with positive GFAP status showed higher conversion risk than negative. In the Cox model, the risk of conversion in the md-SCD group was 2.77 times higher than sd-SCD. The md-SCD group demonstrated a faster rate of Aβ42/40 decline than sd-SCD. CONCLUSIONS The study utilized plasma biomarkers to highlight the significance of staging in SCD. In cognitively normal subjects, md-SCD presents a higher risk of cognitive decline than sd-SCD, providing a valuable reference and convenient tool for early identification of individuals at risk for AD.
Collapse
Affiliation(s)
- Min Wei
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xianfeng Yu
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Shimin Hu
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Wenjing Hu
- Institute of Biomedical Engineering, School of Life Sciences, Shanghai University, Shanghai, China
| | - Rong Shi
- Institute of Biomedical Engineering, School of Life Sciences, Shanghai University, Shanghai, China
| | - Min Wang
- Institute of Biomedical Engineering, School of Life Sciences, Shanghai University, Shanghai, China
| | - Jiayi Zhong
- Institute of Biomedical Engineering, School of Life Sciences, Shanghai University, Shanghai, China
| | - Qi Zhang
- Institute of Biomedical Engineering, School of Life Sciences, Shanghai University, Shanghai, China
| | - Ying Zhang
- Institute of Biomedical Engineering, School of Life Sciences, Shanghai University, Shanghai, China
| | - Chenyang Li
- Institute of Biomedical Engineering, School of Life Sciences, Shanghai University, Shanghai, China
| | - Ziyan Song
- Institute of Biomedical Engineering, School of Life Sciences, Shanghai University, Shanghai, China
| | - Jiehui Jiang
- Institute of Biomedical Engineering, School of Life Sciences, Shanghai University, Shanghai, China
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Hainan, China
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
- National Clinical Research Center for Geriatric Diseases, Beijing, China
- The Central Hospital of Karamay, Xinjiang, China
| |
Collapse
|
4
|
Gibson LL, Gonzalez MC, Ashton NJ, Tovar‐Rios D, Blanc F, Pilotto A, Lemstra A, Paquet C, Ballard C, Zetterberg H, Aarsland D. Plasma phosphorylated tau and neuropsychiatric symptoms in dementia with Lewy bodies. Alzheimers Dement 2025; 21:e14434. [PMID: 39732510 PMCID: PMC11848415 DOI: 10.1002/alz.14434] [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: 07/19/2024] [Revised: 10/18/2024] [Accepted: 11/05/2024] [Indexed: 12/30/2024]
Abstract
INTRODUCTION Neuropsychiatric symptoms (NPSs) are common in dementia with Lewy bodies (DLB) but their neurobiological mechanisms are poorly understood. METHODS NPSs and cognition were assessed annually in participants (DLB n = 222; Alzheimer's disease [AD] n = 125) from the European DLB (E-DLB) Consortium, and plasma phosphorylated tau-181 (p-tau181) and p-tau231 concentrations were measured at baseline. RESULTS Hallucinations, delusions, and depression were more common in DLB than in AD and, in a subgroup with longitudinal follow-up, persistent hallucinations and NPSs were associated with lower p-tau181 and p-tau231 in DLB. In adjusted linear mixed-effects models, hallucinations at baseline were associated with greater longitudinal cognitive impairment in DLB, with a significant interaction with p-tau231. DISCUSSION Higher p-tau181 and p-tau231 levels were associated with a lower longitudinal risk of NPSs and hallucinations in early-stage DLB. However, the interaction between hallucinations and p-tau231 suggests that when AD co-pathology and hallucinations do co-exist in DLB that they may synergistically exacerbate cognitive decline. HIGHLIGHTS Neuropsychiatric symptoms (NPSs) were more common in dementia with Lewy bodies (DLB) than in Alzheimer's disease (AD). Lower plasma phosphorylated tau-231 (p-tau231) and p-tau181 levels were associated with persistent hallucinations in DLB. Lower plasma p-tau231 and p-tau181 levels were associated with an increased risk of persistent NPSs in early DLB. Hallucinations at baseline were associated with greater cognitive dysfunction in DLB, and there was an interaction with p-tau231.
Collapse
Affiliation(s)
- Lucy L. Gibson
- Centre for Healthy Brain AgeingDepartment of Psychological MedicineInstitute of PsychiatryPsychology, and NeuroscienceKing's College LondonLondonUK
| | - Maria C. Gonzalez
- Department of Quality and Health TechnologyFaculty of Health SciencesUniversity of StavangerStavangerNorway
- The Norwegian Centre for Movement DisordersStavanger University HospitalStavangerNorway
- Centre for Age‐Related MedicineStavanger University HospitalStavangerNorway
| | - Nicholas J. Ashton
- Centre for Healthy Brain AgeingDepartment of Psychological MedicineInstitute of PsychiatryPsychology, and NeuroscienceKing's College LondonLondonUK
- Centre for Age‐Related MedicineStavanger University HospitalStavangerNorway
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgGöteborgSweden
- Banner Alzheimer's Institute and University of ArizonaSt PhoenixArizonaUSA
- Banner Sun Health Research InstituteSun CityArizonaUSA
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHSLondonUK
| | - Diego Tovar‐Rios
- Department of Quality and Health TechnologyFaculty of Health SciencesUniversity of StavangerStavangerNorway
| | - Frédéric Blanc
- Memory Resource and Research CentreGeriatrics Day HospitalGeriatrics DepartmentUniversity Hospital of StrasbourgStrasbourgFrance
| | - Andrea Pilotto
- Neurology UnitLaboratory of Digital Neurology and BiosensorsNeurobiorepository and Laboratory of advanced biological markersDepartment of Clinical and Experimental SciencesItaly and Department of continuity of care and frailty, Neurology Unit, ASST Spedali Civili of BresciaUniversity of BresciaBresciaItaly
| | - Afina Lemstra
- Amsterdam Alzheimer CenterAmsterdam University Medical CentersAmsterdamthe Netherlands
| | - Claire Paquet
- Université de ParisNeurology CenterAssistance Publique Hôpitaux de ParisLariboisière Fernand‐Widal HospitalParisFrance
| | - Clive Ballard
- Department of Clinical and Biomedical Sciences, Medical SchoolUniversity of ExeterExeterUK
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgGöteborgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalGöteborgSweden
- Department of Neurodegenerative DiseaseUniveristy College London Institute of NeurologyQueen SquareLondonUK
- UK Dementia Research Institute at University College LondonLondonUK
- Hong Kong Center for Neurodegenerative DiseasesScience ParkHong KongChina
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Dag Aarsland
- Centre for Healthy Brain AgeingDepartment of Psychological MedicineInstitute of PsychiatryPsychology, and NeuroscienceKing's College LondonLondonUK
- Centre for Age‐Related MedicineStavanger University HospitalStavangerNorway
| | | |
Collapse
|
5
|
Anastasi F, Fernández‐Lebrero A, Ashton NJ, Ortiz‐Romero P, Torres‐Torronteras J, González‐Escalante A, Milà‐Alomà M, Contador J, García‐Escobar G, Manero‐Borràs RM, Navalpotro‐Gómez I, Jiménez‐Moyano E, Sahajan A, Hao Q, Zhang B, Jeromin A, Le Bastard N, Nadal A, Mousavi T, Kollmorgen G, Carboni M, Grau‐Rivera O, Zetterberg H, del Campo M, Blennow K, Puig‐Pijoan A, Suárez‐Calvet M. A head-to-head comparison of plasma biomarkers to detect Alzheimer's disease in a memory clinic. Alzheimers Dement 2025; 21:e14609. [PMID: 39998475 PMCID: PMC11852974 DOI: 10.1002/alz.14609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/26/2025]
Abstract
INTRODUCTION Blood-based biomarkers for Alzheimer's disease (AD) have been widely studied, but direct comparisons of several biomarkers in clinical settings remain limited. METHODS In this cross-sectional study, plasma biomarkers from 197 participants in the BIODEGMAR cohort (Hospital del Mar, Barcelona) were analyzed. Participants were classified based on AD cerebrospinal fluid (CSF) core biomarkers. We assessed the ability of plasma p-tau181, p-tau217, p-tau231, t-tau, and Aβ42/40 to classify Aβ pathology status. RESULTS Plasma p-tau biomarkers had a greater diagnostic performance and larger effect sizes compared to t-tau and Aβ42/40 assays in detecting Aβ pathology. Among them, plasma p-tau217 consistently outperformed the others, demonstrating superior area under the curves. Furthermore, p-tau217 showed the strongest correlation between plasma and CSF levels, underscoring its potential as a reliable surrogate for CSF biomarkers. DISCUSSION Several plasma biomarkers, targeting different epitopes and using different platforms, demonstrated high performance in detecting AD in a memory clinic setting. HIGHLIGHTS Plasma p-tau biomarkers demonstrated higher diagnostic performance and larger effect sizes than t-tau and Aβ42/40 assays in detecting Alzheimer's disease. Among the p-tau biomarkers, p-tau217 assays consistently outperformed the others, providing superior classification of Aβ pathology status across different phosphorylation sites. p-tau217 assays showed the strongest correlation between plasma and CSF levels, indicating its potential as a reliable surrogate for CSF biomarkers. Several plasma p-tau biomarkers can be used in a specialized memory clinic to accurately detect Alzheimer's disease.
Collapse
Affiliation(s)
- Federica Anastasi
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Centre for Genomic Regulation (CRG)Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
| | - Aida Fernández‐Lebrero
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
- Department of Medicine and Life SciencesUniversitat Pompeu FabraBarcelonaSpain
| | - Nicholas J. Ashton
- Institute of Neuroscience and Physiology, Department of Psychiatry and NeurochemistryThe Sahlgrenska Academy at University of GothenburgMölndalSweden
- King's College London, Institute of Psychiatry, Psychology & NeuroscienceMaurice Wohl Clinical Neuroscience InstituteLondonUK
- Banner Alzheimer's InstitutePhoenixArizonaUSA
- Banner Sun Health Research InstituteSun CityArizonaUSA
| | - Paula Ortiz‐Romero
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
| | - Javier Torres‐Torronteras
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
| | - Armand González‐Escalante
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Department of Medicine and Life SciencesUniversitat Pompeu FabraBarcelonaSpain
| | - Marta Milà‐Alomà
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - José Contador
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
| | - Greta García‐Escobar
- Hospital del Mar Research InstituteBarcelonaSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
| | | | - Irene Navalpotro‐Gómez
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
- Department of Medicine and Life SciencesUniversitat Pompeu FabraBarcelonaSpain
| | - Esther Jiménez‐Moyano
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
| | | | - Qinyu Hao
- Alamar Biosciences, Inc.FremontCaliforniaUSA
| | | | | | | | | | | | | | | | - Oriol Grau‐Rivera
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)Instituto de Salud Carlos IIIMadridSpain
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and NeurochemistryThe Sahlgrenska Academy at University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water BayHong KongChina
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public Health, University of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Marta del Campo
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de FarmaciaUniversidad San Pablo‐CEU, CEU UniversitiesMadridSpain
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and NeurochemistryThe Sahlgrenska Academy at University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Paris Brain Institute, ICM, Pitié‐Salpêtrière HospitalSorbonne University, 47 Boulevard de l'HôpitalParisFrance
| | - Albert Puig‐Pijoan
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
- Medicine DepartmentUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Marc Suárez‐Calvet
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
| |
Collapse
|
6
|
Teunissen CE, Kolster R, Triana‐Baltzer G, Janelidze S, Zetterberg H, Kolb HC. Plasma p-tau immunoassays in clinical research for Alzheimer's disease. Alzheimers Dement 2025; 21:e14397. [PMID: 39625101 PMCID: PMC11977406 DOI: 10.1002/alz.14397] [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: 04/16/2024] [Revised: 10/04/2024] [Accepted: 10/21/2024] [Indexed: 01/29/2025]
Abstract
The revised biomarker framework for diagnosis and staging of Alzheimer's disease (AD) relies on amyloid beta (Aβ) and tau pathologies as core markers, and markers for adjacent pathophysiology, such as neurodegeneration and inflammation. Many of the core fluid biomarkers are phosphorylated tau (p-tau) fragments, with p-tau217 showing a prominent association with Aβ and tau. While positron emission tomography (PET) imaging is well established, plasma p-tau assays are newer and likely to reduce the use of expensive, and less accessible cerebrospinal fluid and PET imaging tests, thereby promoting wider access to AD screening. There is a need for greater understanding of how the various plasma p-tau species reflect different pathological processes of AD and how different immunoassays perform. This review surveys the available immunoassays and highlights their strengths and limitations in different contexts of use. Assays need to be standardized to maximize their impact on AD clinical research, and patient diagnosis and management. HIGHLIGHTS: Different plasma phosphorylated tau (p-tau) species reflect different pathological processes of Alzheimer's disease (AD), with p-tau231 showing the greatest association with the earliest increases in brain amyloid beta (Aβ) accumulation, while p-tau217 shows greater association with both brain Aβ and early tau pathology, and other p-tau and tau fragment species show greater association with later stages of brain tau pathology. Plasma p-tau217 has proven to be an excellent biomarker for AD pathology due to its close association with both brain Aβ and tau pathology, as well as its large dynamic range. Many different assays with varying performance exist for the same p-tau species, with mass spectrometry assays performing uniformly well, and several immunoassays achieving comparable performance. "Round robin" head-to-head studies have been performed to compare different assays for several key plasma biomarkers, including p-tau181 and p-tau217, but additional head-to-head studies are needed, especially for new analytes and for measuring performance in diverse populations. Plasma immunoassays have the potential to increase accessibility of early diagnostic testing for a broad population, including diverse historically under-represented and under-served populations, due to the potential to be implemented globally, including in primary care settings; however, further research is needed to validate the optimal cutoffs for each assay for real-world clinical usage. Eventually, clinical implementation of a two-step workflow may allow standalone use of plasma testing in certain contexts, minimizing the need for confirmation with costly and less accessible cerebrospinal fluid/positron emission tomography testing.
Collapse
Affiliation(s)
- Charlotte E. Teunissen
- Department of Clinical ChemistryAmsterdam University Medical CentersAmsterdamthe Netherlands
| | | | | | - Shorena Janelidze
- Clinical Memory Research UnitFaculty of MedicineLund UniversityLundSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalGöteborgSweden
- Department of Neurodegenerative DiseaseUCL Institute of Neurology, Queen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | | |
Collapse
|
7
|
An C, Cai H, Ren Z, Fu X, Quan S, Jia L. Biofluid biomarkers for Alzheimer's disease: past, present, and future. MEDICAL REVIEW (2021) 2024; 4:467-491. [PMID: 39664082 PMCID: PMC11629312 DOI: 10.1515/mr-2023-0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 09/04/2024] [Indexed: 12/13/2024]
Abstract
Alzheimer's disease (AD) is a gradually progressive neurodegenerative disease with tremendous social and economic burden. Therefore, early and accurate diagnosis is imperative for effective treatment or prevention of the disease. Cerebrospinal fluid and blood biomarkers emerge as favorable diagnostic tools due to their relative accessibility and potential for widespread clinical use. This review focuses on the AT(N) biomarker system, which includes biomarkers reflecting AD core pathologies, amyloid deposition, and pathological tau, as well as neurodegeneration. Novel biomarkers associated with inflammation/immunity, synaptic dysfunction, vascular pathology, and α-synucleinopathy, which might contribute to either the pathogenesis or the clinical progression of AD, have also been discussed. Other emerging candidates including non-coding RNAs, metabolites, and extracellular vesicle-based markers have also enriched the biofluid biomarker landscape for AD. Moreover, the review discusses the current challenges of biofluid biomarkers in AD diagnosis and offers insights into the prospective future development.
Collapse
Affiliation(s)
- Chengyu An
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Huimin Cai
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Ziye Ren
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Xiaofeng Fu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Shuiyue Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Longfei Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| |
Collapse
|
8
|
Budak M, Fausto BA, Osiecka Z, Sheikh M, Perna R, Ashton N, Blennow K, Zetterberg H, Fitzgerald-Bocarsly P, Gluck MA. Elevated plasma p-tau231 is associated with reduced generalization and medial temporal lobe dynamic network flexibility among healthy older African Americans. Alzheimers Res Ther 2024; 16:253. [PMID: 39578853 PMCID: PMC11583385 DOI: 10.1186/s13195-024-01619-0] [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: 03/21/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Phosphorylated tau (p-tau) and amyloid beta (Aβ) in human plasma may provide an affordable and minimally invasive method to evaluate Alzheimer's disease (AD) pathophysiology. The medial temporal lobe (MTL) is susceptible to changes in structural integrity that are indicative of the disease progression. Among healthy adults, higher dynamic network flexibility within the MTL was shown to mediate better generalization of prior learning, a measure which has been demonstrated to predict cognitive decline and neural changes in preclinical AD longitudinally. Recent developments in cognitive, neural, and blood-based biomarkers of AD risk that may correspond with MTL changes. However, there is no comprehensive study on how these generalization biomarkers, long-term memory, MTL dynamic network flexibility, and plasma biomarkers are interrelated. This study investigated (1) the relationship between long-term memory, generalization performance, and MTL dynamic network flexibility and (2) how plasma p-tau231, p-tau181, and Aβ42/Aβ40 influence generalization, long-term memory, and MTL dynamics in cognitively unimpaired older African Americans. METHODS 148 participants (Meanage: 70.88,SDage: 6.05) were drawn from the ongoing longitudinal study, Pathways to Healthy Aging in African Americans conducted at Rutgers University-Newark. Cognition was evaluated with the Rutgers Acquired Equivalence Task (generalization task) and Rey Auditory Learning Test (RAVLT) delayed recall. MTL dynamic network connectivity was measured from functional Magnetic Resonance Imaging data. Plasma p-tau231, p-tau181, and Aβ42/Aβ40 were measured from blood samples. RESULTS There was a significant positive correlation between generalization performance and MTL Dynamic Network Flexibility (t = 3.372, β = 0.280, p < 0.001). There were significant negative correlations between generalization performance and plasma p-tau231 (t = -3.324, β = -0.265, p = 0.001) and p-tau181 (t = -2.408, β = -0.192, p = 0.017). A significant negative correlation was found between plasma p-tau231 and MTL Dynamic Network Flexibility (t = -2.825, β = -0.232, p = 0.005). CONCLUSIONS Increased levels of p-tau231 are associated with impaired generalization abilities and reduced dynamic network flexibility within the MTL. Plasma p-tau231 may serve as a potential biomarker for assessing cognitive decline and neural changes in cognitively unimpaired older African Americans.
Collapse
Affiliation(s)
- Miray Budak
- Center for Molecular & Behavioral Neuroscience, Rutgers University-Newark, 197 University Avenue, Suite 209, Newark, NJ, 07102, USA.
| | - Bernadette A Fausto
- Center for Molecular & Behavioral Neuroscience, Rutgers University-Newark, 197 University Avenue, Suite 209, Newark, NJ, 07102, USA
| | - Zuzanna Osiecka
- Center for Molecular & Behavioral Neuroscience, Rutgers University-Newark, 197 University Avenue, Suite 209, Newark, NJ, 07102, USA
| | - Mustafa Sheikh
- Center for Molecular & Behavioral Neuroscience, Rutgers University-Newark, 197 University Avenue, Suite 209, Newark, NJ, 07102, USA
| | - Robert Perna
- Center for Molecular & Behavioral Neuroscience, Rutgers University-Newark, 197 University Avenue, Suite 209, Newark, NJ, 07102, USA
| | - Nicholas Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Wallinsgatan 6, Mölndal, Gothenburg, 431 41, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Wallinsgatan 6, Mölndal, Gothenburg, 431 41, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Wallinsgatan 6, Mölndal, Gothenburg, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Box 100, Mölndal, Gothenburg, 405 30, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, 6th Floor, Maple House, Tottenham Ct Rd, London, W1T 7NF, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Units 1501- 1502, 1512-1518, 15/F Building 17W, 17 Science Park W Ave, Science Park, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave J5/1 Mezzanine, Madison, WI, USA
| | - Patricia Fitzgerald-Bocarsly
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Medical Science Building 185 South Orange Avenue, Newark, NJ, USA
| | - Mark A Gluck
- Center for Molecular & Behavioral Neuroscience, Rutgers University-Newark, 197 University Avenue, Suite 209, Newark, NJ, 07102, USA
| |
Collapse
|
9
|
Miner AE, Groh JR, Tripodis Y, Adler CH, Balcer LJ, Bernick C, Zetterberg H, Blennow K, Peskind E, Ashton NJ, Gaudet CE, Martin B, Palmisano JN, Banks SJ, Barr WB, Wethe JV, Cantu RC, Dodick DW, Katz DI, Mez J, van Amerongen S, Cummings JL, Shenton ME, Reiman EM, Stern RA, Alosco ML. Examination of plasma biomarkers of amyloid, tau, neurodegeneration, and neuroinflammation in former elite American football players. Alzheimers Dement 2024; 20:7529-7546. [PMID: 39351900 PMCID: PMC11567811 DOI: 10.1002/alz.14231] [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: 03/14/2024] [Revised: 07/24/2024] [Accepted: 07/27/2024] [Indexed: 11/17/2024]
Abstract
INTRODUCTION Blood-based biomarkers offer a promising approach for the detection of neuropathologies from repetitive head impacts (RHI). We evaluated plasma biomarkers of amyloid, tau, neurodegeneration, and inflammation in former football players. METHODS The sample included 180 former football players and 60 asymptomatic, unexposed male participants (aged 45-74). Plasma assays were conducted for beta-amyloid (Aβ) 40, Aβ42, hyper-phosphorylated tau (p-tau) 181+231, total tau (t-tau), neurofilament light (NfL), glial fibrillary acidic protein (GFAP), interleukin-6 (IL-6), Aβ42/p-tau181 and Aβ42/Aβ40 ratios. We evaluated their ability to differentiate the groups and associations with RHI proxies and traumatic encephalopathy syndrome (TES). RESULTS P-tau181 and p-tau231(padj = 0.016) were higher and Aβ42/p-tau181 was lower(padj = 0.004) in football players compared to controls. Discrimination accuracy for p-tau was modest (area under the curve [AUC] = 0.742). Effects were not attributable to AD-related pathology. Younger age of first exposure (AFE) correlated with higher NfL (padj = 0.03) and GFAP (padj = 0.033). Plasma GFAP was higher in TES-chronic traumatic encephalopathy (TES-CTE) Possible/Probable (padj = 0.008). DISCUSSION Plasma p-tau181 and p-tau231, GFAP, and NfL may offer some usefulness for the characterization of RHI-related neuropathologies. HIGHLIGHTS Former football players had higher plasma p-tau181 and p-tau231 and lower Aβ42/ptau-181 compared to asymptomatic, unexposed men. Younger age of first exposure was associated with increased plasma NfL and GFAP in older but not younger participants. Plasma GFAP was higher in participants with TES-CTE possible/probable compared to TES-CTE no/suggestive.
Collapse
Grants
- ZEN-21-848495 Alzheimer's Association 2021 Zenith Award
- ALZ2022-0006 Hjärnfonden, Sweden
- U01 NS093334 NINDS NIH HHS
- ALFGBG-965240 Hjärnfonden, Sweden
- JPND2021-00694 European Union Joint Programme-Neurodegenerative Disease Research
- UKDRI-1003 UK Dementia Research Institute at UCL
- 2022-00732 UK Dementia Research Institute at UCL
- SG-23-1038904 QC Alzheimer's Association 2022-2025 Grant
- AF-939721 Swedish Alzheimer Foundation
- AF-930351 Swedish Alzheimer Foundation
- RF1 NS132290 NINDS NIH HHS
- AF-994551 Swedish Alzheimer Foundation
- ADSF-21-831381-C AD Strategic Fund and the Alzheimer's Association
- Bluefield Project, Cure Alzheimer's Fund
- JPND2019-466-236 European Union Joint Program for Neurodegenerative Disorders
- 2017-00915 UK Dementia Research Institute at UCL
- Olav Thon Foundation, the Erling-Persson Family Foundation
- FO2017-0243 Hjärnfonden, Sweden
- ADSF-21-831376-C AD Strategic Fund and the Alzheimer's Association
- European Union's Horizon 2020
- ADSF-24-1284328-C AD Strategic Fund and the Alzheimer's Association
- RF1NS132290 National Institute of Neurological Disorders and Stroke/National Institute on Aging
- Kirsten and Freddy Johansen Foundation, Copenhagen, Denmark
- ALFGBG-715986 Hjärnfonden, Sweden
- #ALFGBG-71320 Swedish State Support for Clinical Research
- AF-968270 Swedish Alzheimer Foundation
- ADSF-21-831377-C AD Strategic Fund and the Alzheimer's Association
- FO2022-0270 Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden
- 101053962 European Union's Horizon Europe
- 201809-2016862 Alzheimer Drug Discovery Foundation
- La Fondation Recherche Alzheimer
- U01NS093334 National Institute of Neurological Disorders and Stroke (NINDS)
- National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
Collapse
|
10
|
Barba L, Abu-Rumeileh S, Barthel H, Massa F, Foschi M, Bellomo G, Gaetani L, Thal DR, Parnetti L, Otto M. Clinical and diagnostic implications of Alzheimer's disease copathology in Lewy body disease. Brain 2024; 147:3325-3343. [PMID: 38991041 DOI: 10.1093/brain/awae203] [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: 01/27/2024] [Revised: 05/03/2024] [Accepted: 06/02/2024] [Indexed: 07/13/2024] Open
Abstract
Concomitant Alzheimer's disease (AD) pathology is a frequent event in the context of Lewy body disease (LBD), occurring in approximately half of all cases. Evidence shows that LBD patients with AD copathology show an accelerated disease course, a greater risk of cognitive decline and an overall poorer prognosis. However, LBD-AD cases may show heterogeneous motor and non-motor phenotypes with a higher risk of dementia and, consequently, be not rarely misdiagnosed. In this review, we summarize the current understanding of LBD-AD by discussing the synergistic effects of AD neuropathological changes and Lewy pathology and their clinical relevance. Furthermore, we provide an extensive overview of neuroimaging and fluid biomarkers under assessment for use in LBD-AD and their possible diagnostic and prognostic values. AD pathology can be predicted in vivo by means of CSF, MRI and PET markers, whereas the most promising technique to date for identifying Lewy pathology in different biological tissues is the α-synuclein seed amplification assay. Pathological imaging and CSF AD biomarkers are associated with a higher likelihood of cognitive decline in LBD but do not always mirror the neuropathological severity as in pure AD. Implementing the use of blood-based AD biomarkers might allow faster screening of LBD patients for AD copathology, thus improving the overall diagnostic sensitivity for LBD-AD. Finally, we discuss the literature on novel candidate biomarkers being exploited in LBD-AD to investigate other aspects of neurodegeneration, such as neuroaxonal injury, glial activation and synaptic dysfunction. The thorough characterization of AD copathology in LBD should be taken into account when considering differential diagnoses of dementia syndromes, to allow prognostic evaluation on an individual level, and to guide symptomatic and disease-modifying therapies.
Collapse
Affiliation(s)
- Lorenzo Barba
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle 06120, Germany
| | - Samir Abu-Rumeileh
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle 06120, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig 04103, Germany
| | - Federico Massa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa 16132, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa 16132, Italy
| | - Matteo Foschi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila 67100, Italy
- Department of Neuroscience, Neurology Unit, S. Maria delle Croci Hospital of Ravenna, AUSL Romagna, Ravenna 48121, Italy
| | - Giovanni Bellomo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06129, Italy
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06129, Italy
| | - Dietmar R Thal
- Department of Imaging and Pathology, Laboratory for Neuropathology, Leuven Brain Institute, KU Leuven, Leuven 3001, Belgium
- Department of Pathology, UZ Leuven, Leuven 3000, Belgium
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06129, Italy
| | - Markus Otto
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle 06120, Germany
| |
Collapse
|
11
|
Engels-Domínguez N, Riphagen JM, Van Egroo M, Koops EA, Smegal LF, Becker JA, Prokopiou PC, Bueichekú E, Kwong KK, Rentz DM, Salat DH, Sperling RA, Johnson KA, Jacobs HIL. Lower Locus Coeruleus Integrity Signals Elevated Entorhinal Tau and Clinical Progression in Asymptomatic Older Individuals. Ann Neurol 2024; 96:650-661. [PMID: 39007398 PMCID: PMC11534559 DOI: 10.1002/ana.27022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 07/16/2024]
Abstract
OBJECTIVE Elevated entorhinal cortex (EC) tau in low beta-amyloid individuals can predict accumulation of pathology and cognitive decline. We compared the accuracy of magnetic resonance imaging (MRI)-derived locus coeruleus integrity, neocortical beta-amyloid burden by positron emission tomography (PET), and hippocampal volume in identifying elevated entorhinal tau signal in asymptomatic individuals who are considered beta-amyloid PET-negative. METHODS We included 188 asymptomatic individuals (70.78 ± 11.51 years, 58% female) who underwent 3T-MRI of the locus coeruleus, Pittsburgh compound-B (PiB), and Flortaucipir (FTP) PET. Associations between elevated EC tau and neocortical PiB, hippocampal volume, or locus coeruleus integrity were evaluated and compared using logistic regression and receiver operating characteristic analyses in the PiB- sample with a clinical dementia rating (CDR) of 0. Associations with clinical progression (CDR-sum-of-boxes) over a time span of 6 years were evaluated with Cox proportional hazard models. RESULTS We identified 26 (21%) individuals with high EC FTP in the CDR = 0/PiB- sample. Locus coeruleus integrity was a significantly more sensitive and specific predictor of elevated EC FTP (area under the curve [AUC] = 85%) compared with PiB (AUC = 77%) or hippocampal volume (AUC = 76%). Based on the Youden-index, locus coeruleus integrity obtained a sensitivity of 77% and 85% specificity. Using the resulting locus coeruleus Youden cut-off, lower locus coeruleus integrity was associated with a two-fold increase in clinical progression, including mild cognitive impairment. INTERPRETATION Locus coeruleus integrity has promise as a low-cost, non-invasive screening instrument to detect early cortical tau deposition and associated clinical progression in asymptomatic, low beta-amyloid individuals. ANN NEUROL 2024;96:650-661.
Collapse
Affiliation(s)
- Nina Engels-Domínguez
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Joost M Riphagen
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Maxime Van Egroo
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Elouise A Koops
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Lindsay F Smegal
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - J Alex Becker
- Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Prokopis C Prokopiou
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Elisenda Bueichekú
- Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Kenneth K Kwong
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Dorene M Rentz
- Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - David H Salat
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Reisa A Sperling
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Keith A Johnson
- Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Heidi I L Jacobs
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
12
|
Kwon HS, Yu HJ, Koh SH. Revolutionizing Alzheimer's Diagnosis and Management: The Dawn of Biomarker-Based Precision Medicine. Dement Neurocogn Disord 2024; 23:188-201. [PMID: 39512700 PMCID: PMC11538857 DOI: 10.12779/dnd.2024.23.4.188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/15/2024] Open
Abstract
Alzheimer's disease (AD), a leading cause of dementia, presents a formidable global health challenge intensified by the aging population. This review encapsulates the evolving landscape of AD diagnosis and treatment with a special focus on the innovative role of fluid biomarkers. Pathologically, AD is marked by amyloid beta (Aβ) plaques and neurofibrillary tangles of hyperphosphorylated tau, which lead to synaptic dysfunction, neuronal loss, and cognitive decline. These pathological changes, commencing decades before symptom onset, underscore the need for early detection and intervention. Diagnosis traditionally relies on clinical assessment, neuropsychological testing, and neuroimaging techniques. However, fluid biomarkers in cerebrospinal fluid and blood, such as various forms of Aβ, total tau, phosphorylated tau, and neurofilament light chain, are emerging as less invasive, cost-effective diagnostic tools. These biomarkers are pivotal for early diagnosis, differential diagnosis, disease progression monitoring, and treatment response evaluation. The treatment landscape is shifting toward personalized medicine, highlighted by advancements in Aβ immunotherapies, such as lecanemab and donanemab. Demonstrating efficacy in phase III clinical trials, these therapies hold promise as tailored treatment strategies based on individual biomarker profiles. The integration of fluid biomarkers into clinical practice represents a significant advance in AD management, providing the potential for early and precise diagnosis, coupled with personalized therapeutic approaches. This heralds a new era in combating this debilitating disease.
Collapse
Affiliation(s)
- Hyuk Sung Kwon
- Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
| | - Hyun-Jung Yu
- Department of Neurology, Bundang Jesaeng General Hospital, Seongnam, Korea
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, Korea
| |
Collapse
|
13
|
Guo T, Li A, Sun P, He Z, Cai Y, Lan G, Liu L, Li J, Yang J, Zhu Y, Zhao R, Chen X, Shi D, Liu Z, Wang Q, Xu L, Zhou L, Ran P, Wang X, Sun K, Lu J, Han Y. Astrocyte reactivity is associated with tau tangle load and cortical thinning in Alzheimer's disease. Mol Neurodegener 2024; 19:58. [PMID: 39080744 PMCID: PMC11290175 DOI: 10.1186/s13024-024-00750-8] [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: 01/15/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND It is not fully established whether plasma β-amyloid(Aβ)42/Aβ40 and phosphorylated Tau181 (p-Tau181) can effectively detect Alzheimer's disease (AD) pathophysiology in older Chinese adults and how these biomarkers correlate with astrocyte reactivity, Aβ plaque deposition, tau tangle aggregation, and neurodegeneration. METHODS We recruited 470 older adults and analyzed plasma Aβ42/Aβ40, p-Tau181, glial fibrillary acidic protein (GFAP), and neurofilament light (NfL) using the Simoa platform. Among them, 301, 195, and 70 underwent magnetic resonance imaging, Aβ and tau positron emission tomography imaging. The plasma Aβ42/Aβ40 and p-Tau181 thresholds were defined as ≤0.0609 and ≥2.418 based on the receiver operating characteristic curve analysis using the Youden index by comparing Aβ-PET negative cognitively unimpaired individuals and Aβ-PET positive cognitively impaired patients. To evaluate the feasibility of using plasma Aβ42/Aβ40 (A) and p-Tau181 (T) to detect AD and understand how astrocyte reactivity affects this process, we compared plasma GFAP, Aβ plaque, tau tangle, plasma NfL, hippocampal volume, and temporal-metaROI cortical thickness between different plasma A/T profiles and explored their relations with each other using general linear models, including age, sex, APOE-ε4, and diagnosis as covariates. RESULTS Plasma A+/T + individuals showed the highest levels of astrocyte reactivity, Aβ plaque, tau tangle, and axonal degeneration, and the lowest hippocampal volume and temporal-metaROI cortical thickness. Lower plasma Aβ42/Aβ40 and higher plasma p-Tau181 were independently and synergistically correlated with higher plasma GFAP and Aβ plaque. Elevated plasma p-Tau181 and GFAP concentrations were directly and interactively associated with more tau tangle formation. Regarding neurodegeneration, higher plasma p-Tau181 and GFAP concentrations strongly correlated with more axonal degeneration, as measured by plasma NfL, and lower plasma Aβ42/Aβ40 and higher plasma p-Tau181 were related to greater hippocampal atrophy. Higher plasma GFAP levels were associated with thinner cortical thickness and significantly interacted with lower plasma Aβ42/Aβ40 and higher plasma p-Tau181 in predicting more temporal-metaROI cortical thinning. Voxel-wise imaging analysis confirmed these findings. DISCUSSION This study provides a valuable reference for using plasma biomarkers to detect AD in the Chinese community population and offers novel insights into how astrocyte reactivity contributes to AD progression, highlighting the importance of targeting reactive astrogliosis to prevent AD.
Collapse
Affiliation(s)
- Tengfei Guo
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China.
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Anqi Li
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Pan Sun
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Zhengbo He
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Yue Cai
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Guoyu Lan
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Lin Liu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Jieyin Li
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Jie Yang
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
- Department of Neurology, Xuanwu Hospital of Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Yalin Zhu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Ruiyue Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510120, China
| | - Xuhui Chen
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, 518000, China
| | - Dai Shi
- Neurology Medicine Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, China
| | - Zhen Liu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Qingyong Wang
- Department of Neurology, Shenzhen Guangming District People's Hospital, Shenzhen, 518107, China
| | - Linsen Xu
- Department of Medical Imaging, Shenzhen Guangming District People's Hospital, Shenzhen, 518106, China
| | - Liemin Zhou
- Neurology Medicine Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, China
| | - Pengcheng Ran
- Department of Nuclear Medicine, Guangdong Hospital of Traditional Chinese Medicine, Guangzhou, 510120, China
| | - Xinlu Wang
- Department of Nuclear Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510120, China
| | - Kun Sun
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Jie Lu
- Department of Neurology, Xuanwu Hospital of Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China.
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China.
| | - Ying Han
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China.
- Department of Neurology, Xuanwu Hospital of Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China.
- School of Biomedical Engineering, Hainan University, Haikou, 570228, China.
- Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, 100053, China.
- National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China.
| |
Collapse
|
14
|
Zeng X, Chen Y, Sehrawat A, Lee J, Lafferty TK, Kofler J, Berman SB, Sweet RA, Tudorascu DL, Klunk WE, Ikonomovic MD, Pfister A, Zetterberg H, Snitz BE, Cohen AD, Villemagne VL, Pascoal TA, Kamboh ML, Lopez OI, Blennow K, Karikari TK. Alzheimer blood biomarkers: practical guidelines for study design, sample collection, processing, biobanking, measurement and result reporting. Mol Neurodegener 2024; 19:40. [PMID: 38750570 PMCID: PMC11095038 DOI: 10.1186/s13024-024-00711-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 02/13/2024] [Indexed: 05/19/2024] Open
Abstract
Alzheimer's disease (AD), the most common form of dementia, remains challenging to understand and treat despite decades of research and clinical investigation. This might be partly due to a lack of widely available and cost-effective modalities for diagnosis and prognosis. Recently, the blood-based AD biomarker field has seen significant progress driven by technological advances, mainly improved analytical sensitivity and precision of the assays and measurement platforms. Several blood-based biomarkers have shown high potential for accurately detecting AD pathophysiology. As a result, there has been considerable interest in applying these biomarkers for diagnosis and prognosis, as surrogate metrics to investigate the impact of various covariates on AD pathophysiology and to accelerate AD therapeutic trials and monitor treatment effects. However, the lack of standardization of how blood samples and collected, processed, stored analyzed and reported can affect the reproducibility of these biomarker measurements, potentially hindering progress toward their widespread use in clinical and research settings. To help address these issues, we provide fundamental guidelines developed according to recent research findings on the impact of sample handling on blood biomarker measurements. These guidelines cover important considerations including study design, blood collection, blood processing, biobanking, biomarker measurement, and result reporting. Furthermore, the proposed guidelines include best practices for appropriate blood handling procedures for genetic and ribonucleic acid analyses. While we focus on the key blood-based AD biomarkers for the AT(N) criteria (e.g., amyloid-beta [Aβ]40, Aβ42, Aβ42/40 ratio, total-tau, phosphorylated-tau, neurofilament light chain, brain-derived tau and glial fibrillary acidic protein), we anticipate that these guidelines will generally be applicable to other types of blood biomarkers. We also anticipate that these guidelines will assist investigators in planning and executing biomarker research, enabling harmonization of sample handling to improve comparability across studies.
Collapse
Affiliation(s)
- Xuemei Zeng
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Yijun Chen
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Anuradha Sehrawat
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Jihui Lee
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Tara K Lafferty
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Julia Kofler
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Sarah B Berman
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Robert A Sweet
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Dana L Tudorascu
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - William E Klunk
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Milos D Ikonomovic
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh HS, Pittsburgh, PA, USA
| | - Anna Pfister
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Beth E Snitz
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Anne D Cohen
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Victor L Villemagne
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Tharick A Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - M. llyas Kamboh
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Oscar I Lopez
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Thomas K Karikari
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA.
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.
| |
Collapse
|
15
|
Kong Q, Liu C, Zhang Y, He Y, Zhang R, Wang Y, Zhou Q, Cui F. Nucleic acid aptamer-based electrochemical sensor for the detection of serum P-tau231 and the instant screening test of Alzheimer's disease. Mikrochim Acta 2024; 191:328. [PMID: 38743383 DOI: 10.1007/s00604-024-06395-0] [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: 01/14/2024] [Accepted: 04/27/2024] [Indexed: 05/16/2024]
Abstract
The instant screening of patients with a tendency towards developing Alzheimer's disease (AD) is significant for providing preventive measures and treatment. However, the current imaging-based technology cannot meet the requirements in the early stage. Developing biosensor-based liquid biopsy technology could be overcoming this bottleneck problem. Herein, we developed a simple, low-cost, and sensitive electrochemical aptamer biosensor for detecting phosphorylated tau protein threonine 231 (P-tau231), the earliest and one of the most efficacious abnormally elevated biomarkers of AD. Gold nanoparticles (AuNPs) were electrochemically synthesized on a glassy carbon electrode as the transducer, exhibiting excellent conductivity, and were applied to amplify the electrochemical signal. A nucleic acid aptamer was designed as the receptor to capture the P-tau231 protein, specifically through the formation of an aptamer-antigen complex. The proposed biosensor showed excellent sensitivity in detecting P-tau 231, with a broad linear detection range from 10 to 107 pg/mL and a limit of detection (LOD) of 2.31 pg/mL. The recoveries of the biosensor in human serum ranged from 97.59 to 103.26%, demonstrating that the biosensor could be used in complex practical samples. In addition, the results showed that the developed biosensor has good repeatability, reproducibility, and stability, which provides a novel method for the early screening of AD.
Collapse
Affiliation(s)
- Qingfei Kong
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China
- The Heilongjiang Provincial Joint Laboratory of Basic Medicine and Multiple Organ System Diseases (International Cooperation), Harbin, Heilongjiang, 150086, China
| | - Chunhan Liu
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Yanlin Zhang
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Yifan He
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Ruiting Zhang
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Yuhan Wang
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Qin Zhou
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China.
- The Heilongjiang Provincial Joint Laboratory of Basic Medicine and Multiple Organ System Diseases (International Cooperation), Harbin, Heilongjiang, 150086, China.
| | - Feiyun Cui
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China.
- The Heilongjiang Provincial Joint Laboratory of Basic Medicine and Multiple Organ System Diseases (International Cooperation), Harbin, Heilongjiang, 150086, China.
| |
Collapse
|
16
|
Yakoub Y, Gonzalez-Ortiz F, Ashton NJ, Déry C, Strikwerda-Brown C, St-Onge F, Ourry V, Schöll M, Geddes MR, Ducharme S, Montembeault M, Rosa-Neto P, Soucy JP, Breitner JCS, Zetterberg H, Blennow K, Poirier J, Villeneuve S. Plasma p-tau217 predicts cognitive impairments up to ten years before onset in normal older adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.09.24307120. [PMID: 38766113 PMCID: PMC11100946 DOI: 10.1101/2024.05.09.24307120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Importance Positron emission tomography (PET) biomarkers are the gold standard for detection of Alzheimer amyloid and tau in vivo . Such imaging can identify cognitively unimpaired (CU) individuals who will subsequently develop cognitive impartment (CI). Plasma biomarkers would be more practical than PET or even cerebrospinal fluid (CSF) assays in clinical settings. Objective Assess the prognostic accuracy of plasma p-tau217 in comparison to CSF and PET biomarkers for predicting the clinical progression from CU to CI. Design In a cohort of elderly at high risk of developing Alzheimer's dementia (AD), we measured the proportion of CU individuals who developed CI, as predicted by Aβ (A+) and/or tau (T+) biomarker assessment from plasma, CSF, and PET. Results from each method were compared with (A-T-) reference individuals. Data were analyzed from June 2023 to April 2024. Setting Longitudinal observational cohort. Participants Some 228 participants from the PREVENT-AD cohort were CU at the time of biomarker assessment and had 1 - 10 years of follow-up. Plasma was available from 215 participants, CSF from 159, and amyloid- and tau-PET from 155. Ninety-three participants had assessment using all three methods (main group of interest). Progression to CI was determined by clinical consensus among physicians and neuropsychologists who were blind to plasma, CSF, PET, and MRI findings, as well as APOE genotype. Exposures Plasma Aβ 42/40 was measured using IP-MS; CSF Aβ 42/40 using Lumipulse; plasma and CSF p-tau217 using UGOT assay. Aβ-PET employed the 18 F-NAV4694 ligand, and tau-PET used 18 F-flortaucipir. Main Outcome Prognostic accuracy of plasma, CSF, and PET biomarkers for predicting the development of CI in CU individuals. Results Cox proportional hazard models indicated a greater progression rate in all A+T+ groups compared to A-T-groups (HR = 6.61 [95% CI = 2.06 - 21.17] for plasma, 3.62 [1.49 - 8.81] for CSF and 9.24 [2.34 - 36.43] for PET). The A-T+ groups were small, but also characterized with individuals who developed CI. Plasma biomarkers identified about five times more T+ than PET. Conclusion and relevance Plasma p-tau217 assessment is a practical method for identification of persons who will develop cognitive impairment up to 10 years later. Key Points Question: Can plasma p-tau217 serve as a prognostic indicator for identifying cognitively unimpaired (CU) individuals at risk of developing cognitive impairments (CI)?Findings: In a longitudinal cohort of CU individuals with a family history of sporadic AD, almost all individuals with abnormal plasma p-tau217 concentrations developed CI within 10 years, regardless of plasma amyloid levels. Similar findings were obtained with CSF p-tau217 and tau-PET. Fluid p-tau217 biomarkers had the main advantage over PET of identifying five times more participants with elevated tau.Meaning: Elevated plasma p-tau217 levels in CU individuals strongly indicate future clinical progression.
Collapse
|
17
|
Mantellatto Grigoli M, Pelegrini LNC, Whelan R, Cominetti MR. Present and Future of Blood-Based Biomarkers of Alzheimer's Disease: Beyond the Classics. Brain Res 2024; 1830:148812. [PMID: 38369085 DOI: 10.1016/j.brainres.2024.148812] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/13/2023] [Accepted: 02/13/2024] [Indexed: 02/20/2024]
Abstract
The field of blood-based biomarkers for Alzheimer's disease (AD) has advanced at an incredible pace, especially after the development of sensitive analytic platforms that can facilitate large-scale screening. Such screening will be important when more sophisticated diagnostic methods are scarce and expensive. Thus, blood-based biomarkers can potentially reduce diagnosis inequities among populations from different socioeconomic contexts. This large-scale screening can be performed so that older adults at risk of cognitive decline assessed using these methods can then undergo more complete assessments with classic biomarkers, increasing diagnosis efficiency and reducing costs to the health systems. Blood-based biomarkers can also aid in assessing the effect of new disease-modifying treatments. This paper reviews recent advances in the area, focusing on the following leading candidates for blood-based biomarkers: amyloid-beta (Aβ), phosphorylated tau isoforms (p-tau), neurofilament light (NfL), and glial fibrillary acidic (GFAP) proteins, as well as on new candidates, Neuron-Derived Exosomes contents (NDEs) and Transactive response DNA-binding protein-43 (TDP-43), based on data from longitudinal observational cohort studies. The underlying challenges of validating and incorporating these biomarkers into routine clinical practice and primary care settings are also discussed. Importantly, challenges related to the underrepresentation of ethnic minorities and socioeconomically disadvantaged persons must be considered. If these challenges are overcome, a new time of cost-effective blood-based biomarkers for AD could represent the future of clinical procedures in the field and, together with continued prevention strategies, the beginning of an era with a lower incidence of dementia worldwide.
Collapse
Affiliation(s)
| | | | - Robert Whelan
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland; School of Psychology, Trinity College Dublin, Dublin, Ireland
| | - Marcia R Cominetti
- Department of Gerontology, Federal University of São Carlos, Brazil; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
18
|
Chen A, Shea D, Daggett V. Performance of SOBA-AD blood test in discriminating Alzheimer's disease patients from cognitively unimpaired controls in two independent cohorts. Sci Rep 2024; 14:7946. [PMID: 38575622 PMCID: PMC10995183 DOI: 10.1038/s41598-024-57107-w] [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: 01/02/2024] [Accepted: 03/14/2024] [Indexed: 04/06/2024] Open
Abstract
Amyloid-beta (Aβ) toxic oligomers are critical early players in the molecular pathology of Alzheimer's disease (AD). We have developed a Soluble Oligomer Binding Assay (SOBA-AD) for detection of these Aβ oligomers that contain α-sheet secondary structure that discriminates plasma samples from patients on the AD continuum from non-AD controls. We tested 265 plasma samples from two independent cohorts to investigate the performance of SOBA-AD. Testing was performed at two different sites, with different personnel, reagents, and instrumentation. Across two cohorts, SOBA-AD discriminated AD patients from cognitively unimpaired (CU) subjects with 100% sensitivity, > 95% specificity, and > 98% area under the curve (AUC) (95% CI 0.95-1.00). A SOBA-AD positive readout, reflecting α-sheet toxic oligomer burden, was found in AD patients, and not in controls, providing separation of the two populations, aside from 5 SOBA-AD positive controls. Based on an earlier SOBA-AD study, the Aβ oligomers detected in these CU subjects may represent preclinical cases of AD. The results presented here support the value of SOBA-AD as a promising blood-based tool for the detection and confirmation of AD.
Collapse
Affiliation(s)
- Amy Chen
- AltPep Corporation, 1150 Eastlake Avenue N, Suite 800, Seattle, WA, 98109, USA
| | - Dylan Shea
- AltPep Corporation, 1150 Eastlake Avenue N, Suite 800, Seattle, WA, 98109, USA
- University of Washington, Box 355610, Seattle, WA, 98195-5610, USA
| | - Valerie Daggett
- AltPep Corporation, 1150 Eastlake Avenue N, Suite 800, Seattle, WA, 98109, USA.
- University of Washington, Box 355610, Seattle, WA, 98195-5610, USA.
| |
Collapse
|
19
|
De Meyer S, Blujdea ER, Schaeverbeke J, Reinartz M, Luckett ES, Adamczuk K, Van Laere K, Dupont P, Teunissen CE, Vandenberghe R, Poesen K. Longitudinal associations of serum biomarkers with early cognitive, amyloid and grey matter changes. Brain 2024; 147:936-948. [PMID: 37787146 DOI: 10.1093/brain/awad330] [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: 04/01/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 10/04/2023] Open
Abstract
Blood-based biomarkers have been extensively evaluated for their diagnostic potential in Alzheimer's disease. However, their relative prognostic and monitoring capabilities for cognitive decline, amyloid-β (Aβ) accumulation and grey matter loss in cognitively unimpaired elderly require further investigation over extended time periods. This prospective cohort study in cognitively unimpaired elderly [n = 185, mean age (range) = 69 (53-84) years, 48% female] examined the prognostic and monitoring capabilities of glial fibrillary acidic protein (GFAP), neurofilament light (NfL), Aβ1-42/Aβ1-40 and phosphorylated tau (pTau)181 through their quantification in serum. All participants underwent baseline Aβ-PET, MRI and blood sampling as well as 2-yearly cognitive testing. A subset additionally underwent Aβ-PET (n = 109), MRI (n = 106) and blood sampling (n = 110) during follow-up [median time interval (range) = 6.1 (1.3-11.0) years]. Matching plasma measurements were available for Aβ1-42/Aβ1-40 and pTau181 (both n = 140). Linear mixed-effects models showed that high serum GFAP and NfL predicted future cognitive decline in memory (βGFAP×Time = -0.021, PFDR = 0.007 and βNfL×Time = -0.031, PFDR = 0.002) and language (βGFAP×Time = -0.021, PFDR = 0.002 and βNfL×Time = -0.018, PFDR = 0.03) domains. Low serum Aβ1-42/Aβ1-40 equally but independently predicted memory decline (βAβ1-42/Aβ1-40×Time = -0.024, PFDR = 0.02). Whole-brain voxelwise analyses revealed that low Aβ1-42/Aβ1-40 predicted Aβ accumulation within the precuneus and frontal regions, high GFAP and NfL predicted grey matter loss within hippocampal regions and low Aβ1-42/Aβ1-40 predicted grey matter loss in lateral temporal regions. Serum GFAP, NfL and pTau181 increased over time, while Aβ1-42/Aβ1-40 decreased only in Aβ-PET-negative elderly. NfL increases associated with declining memory (βNfLchange×Time = -0.030, PFDR = 0.006) and language (βNfLchange×Time = -0.021, PFDR = 0.02) function and serum Aβ1-42/Aβ1-40 decreases associated with declining language function (βAβ1-42/Aβ1-40×Time = -0.020, PFDR = 0.04). GFAP increases associated with Aβ accumulation within the precuneus and NfL increases associated with grey matter loss. Baseline and longitudinal serum pTau181 only associated with Aβ accumulation in restricted occipital regions. In head-to-head comparisons, serum outperformed plasma Aβ1-42/Aβ1-40 (ΔAUC = 0.10, PDeLong, FDR = 0.04), while both plasma and serum pTau181 demonstrated poor performance to detect asymptomatic Aβ-PET positivity (AUC = 0.55 and 0.63, respectively). However, when measured with a more phospho-specific assay, plasma pTau181 detected Aβ-positivity with high performance (AUC = 0.82, PDeLong, FDR < 0.007). In conclusion, serum GFAP, NfL and Aβ1-42/Aβ1-40 are valuable prognostic and/or monitoring tools in asymptomatic stages providing complementary information in a time- and pathology-dependent manner.
Collapse
Affiliation(s)
- Steffi De Meyer
- Laboratory for Molecular Neurobiomarker Research, Department of Neurosciences, KU Leuven, 3000 Leuven, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, 3000 Leuven, Belgium
- Alzheimer Research Centre, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
| | - Elena R Blujdea
- Neurochemistry Laboratory, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Jolien Schaeverbeke
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, 3000 Leuven, Belgium
- Alzheimer Research Centre, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
| | - Mariska Reinartz
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, 3000 Leuven, Belgium
- Alzheimer Research Centre, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
| | - Emma S Luckett
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, 3000 Leuven, Belgium
- Alzheimer Research Centre, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
| | - Katarzyna Adamczuk
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, 3000 Leuven, Belgium
| | - Koen Van Laere
- Alzheimer Research Centre, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
- Division of Nuclear Medicine, UZ Leuven, 3000 Leuven, Belgium
| | - Patrick Dupont
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, 3000 Leuven, Belgium
- Alzheimer Research Centre, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
| | | | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, 3000 Leuven, Belgium
- Alzheimer Research Centre, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
- Department of Neurology, UZ Leuven, 3000 Leuven, Belgium
| | - Koen Poesen
- Laboratory for Molecular Neurobiomarker Research, Department of Neurosciences, KU Leuven, 3000 Leuven, Belgium
- Alzheimer Research Centre, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
- Department of Laboratory Medicine, UZ Leuven, 3000 Leuven, Belgium
| |
Collapse
|
20
|
Jack CR, Wiste HJ, Algeciras‐Schimnich A, Weigand SD, Figdore DJ, Lowe VJ, Vemuri P, Graff‐Radford J, Ramanan VK, Knopman DS, Mielke MM, Machulda MM, Fields J, Schwarz CG, Cogswell PM, Senjem ML, Therneau TM, Petersen RC. Comparison of plasma biomarkers and amyloid PET for predicting memory decline in cognitively unimpaired individuals. Alzheimers Dement 2024; 20:2143-2154. [PMID: 38265198 PMCID: PMC10984437 DOI: 10.1002/alz.13651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND We compared the ability of several plasma biomarkers versus amyloid positron emission tomography (PET) to predict rates of memory decline among cognitively unimpaired individuals. METHODS We studied 645 Mayo Clinic Study of Aging participants. Predictor variables were age, sex, education, apolipoprotein E (APOE) ε4 genotype, amyloid PET, and plasma amyloid beta (Aβ)42/40, phosphorylated tau (p-tau)181, neurofilament light (NfL), glial fibrillary acidic protein (GFAP), and p-tau217. The outcome was a change in a memory composite measure. RESULTS All plasma biomarkers, except NfL, were associated with mean memory decline in models with individual biomarkers. However, amyloid PET and plasma p-tau217, along with age, were key variables independently associated with mean memory decline in models combining all predictors. Confidence intervals were narrow for estimates of population mean prediction, but person-level prediction intervals were wide. DISCUSSION Plasma p-tau217 and amyloid PET provide useful information about predicting rates of future cognitive decline in cognitively unimpaired individuals at the population mean level, but not at the individual person level.
Collapse
Affiliation(s)
| | - Heather J. Wiste
- Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | | | - Stephen D. Weigand
- Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | - Dan J. Figdore
- Department of Laboratory MedicineMayo ClinicRochesterMinnesotaUSA
| | - Val J. Lowe
- Department of Nuclear MedicineMayo ClinicRochesterMinnesotaUSA
| | | | | | | | | | - Michelle M. Mielke
- Department of Epidemiology and PreventionWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Mary M. Machulda
- Department of Psychiatry and PsychologyMayo ClinicRochesterMinnesotaUSA
| | - Julie Fields
- Department of Psychiatry and PsychologyMayo ClinicRochesterMinnesotaUSA
| | | | | | | | - Terry M. Therneau
- Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | | |
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
|
Cogswell PM, Lundt ES, Therneau TM, Wiste HJ, Graff‐Radford J, Algeciras‐Schimnich A, Lowe VJ, Mielke MM, Schwarz CG, Senjem ML, Gunter JL, Knopman DS, Vemuri P, Petersen RC, Jack Jr CR. Modeling the temporal evolution of plasma p-tau in relation to amyloid beta and tau PET. Alzheimers Dement 2024; 20:1225-1238. [PMID: 37963289 PMCID: PMC10916944 DOI: 10.1002/alz.13539] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/02/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023]
Abstract
INTRODUCTION The timing of plasma biomarker changes is not well understood. The goal of this study was to evaluate the temporal co-evolution of plasma and positron emission tomography (PET) Alzheimer's disease (AD) biomarkers. METHODS We included 1408 Mayo Clinic Study of Aging and Alzheimer's Disease Research Center participants. An accelerated failure time (AFT) model was fit with amyloid beta (Aβ) PET, tau PET, plasma p-tau217, p-tau181, and glial fibrillary acidic protein (GFAP) as endpoints. RESULTS Individual timing of plasma p-tau progression was strongly associated with Aβ PET and GFAP progression. In the population, GFAP became abnormal first, then Aβ PET, plasma p-tau, and tau PET temporal meta-regions of interest when applying cut points based on young, cognitively unimpaired participants. DISCUSSION Plasma p-tau is a stronger indicator of a temporally linked response to elevated brain Aβ than of tau pathology. While Aβ deposition and a rise in GFAP are upstream events associated with tau phosphorylation, the temporal link between p-tau and Aβ PET was the strongest. HIGHLIGHTS Plasma p-tau progression was more strongly associated with Aβ than tau PET. Progression on plasma p-tau was associated with Aβ PET and GFAP progression. P-tau181 and p-tau217 become abnormal after Aβ PET and before tau PET. GFAP became abnormal first, before plasma p-tau and Aβ PET.
Collapse
Affiliation(s)
| | - Emily S. Lundt
- Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | - Terry M. Therneau
- Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | - Heather J. Wiste
- Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | | | | | - Val J. Lowe
- Department of RadiologyMayo ClinicRochesterMinnesotaUSA
| | - Michelle M. Mielke
- Department of Epidemiology and PreventionWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | | | - Matthew L. Senjem
- Department of RadiologyMayo ClinicRochesterMinnesotaUSA
- Department of Information TechnologyMayo ClinicRochesterMinnesotaUSA
| | | | | | | | - Ronald C. Petersen
- Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
| | | |
Collapse
|
23
|
Li Z, Fan Z, Zhang Q. The Associations of Phosphorylated Tau 181 and Tau 231 Levels in Plasma and Cerebrospinal Fluid with Cognitive Function in Alzheimer's Disease: A Systematic Review and Meta-Analysis. J Alzheimers Dis 2024; 98:13-32. [PMID: 38339929 DOI: 10.3233/jad-230799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Background Cerebrospinal fluid (CSF) or blood biomarkers like phosphorylated tau proteins (p-tau) are used to detect Alzheimer's disease (AD) early. Increasing studies on cognitive function and blood or CSF p-tau levels are controversial. Objective Our study examined the potential of p-tau as a biomarker of cognitive status in normal control (NC), mild cognitive impairment (MCI), and AD patients. Methods We searched PubMed, Cochrane, Embase, and Web of Science for relevant material through 12 January 2023. 5,017 participants from 20 studies-1,033 AD, 2,077 MCI, and 1,907 NC-were evaluated. Quantitative analysis provided continuous outcomes as SMDs with 95% CIs. Begg tested publication bias. Results MCI patients had lower CSF p-tau181 levels than AD patients (SMD =-0.60, 95% CI (-0.85, -0.36)) but higher than healthy controls (SMD = 0.67). AD/MCI patients had greater plasma p-tau181 levels than healthy people (SMD =-0.73, 95% CI (-1.04, -0.43)). MCI patients had significantly lower p-tau231 levels than AD patients in plasma and CSF (SMD =-0.90, 95% CI (-0.82, -0.45)). MCI patients showed greater CSF and plasma p-tau231 than healthy controls (SMD = 1.34, 95% CI (0.89, 1.79) and 0.43, (0.23, 0.64)). Plasma p-tau181/231 levels also distinguished the three categories. MCI patients had higher levels than healthy people, while AD patients had higher levels than MCI patients. Conclusions CSF p-tau181 and p-tau231 biomarkers distinguished AD, MCI, and healthy populations. Plasma-based p-tau181 and p-tau231 biomarkers for AD and MCI need further study.
Collapse
Affiliation(s)
- Zhirui Li
- Department of Disease Control and Prevention, Sichuan Provincial Center for Disease Control and Prevention, Sichuan Chengdu, China
| | - Zixuan Fan
- School of Health Policy and Management, Peking Union Medical College, Beijing, China
| | - Qian Zhang
- Department of Oncology, Xiamen Fifth Hospital, Fujian Xiamen, China
| |
Collapse
|
24
|
Yakoub Y, Ashton NJ, Strikwerda-Brown C, Montoliu-Gaya L, Karikari TK, Kac PR, Gonzalez-Ortiz F, Gallego-Rudolf J, Meyer PF, St-Onge F, Schöll M, Soucy JP, Breitner JCS, Zetterberg H, Blennow K, Poirier J, Villeneuve S. Longitudinal blood biomarker trajectories in preclinical Alzheimer's disease. Alzheimers Dement 2023; 19:5620-5631. [PMID: 37294682 DOI: 10.1002/alz.13318] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/03/2023] [Accepted: 05/11/2023] [Indexed: 06/11/2023]
Abstract
INTRODUCTION Plasma biomarkers are altered years prior to Alzheimer's disease (AD) clinical onset. METHODS We measured longitudinal changes in plasma amyloid-beta (Aβ)42/40 ratio, pTau181, pTau231, neurofilament light chain (NfL), and glial fibrillary acidic protein (GFAP) in a cohort of older adults at risk of AD (n = 373 total, n = 229 with Aβ and tau positron emission tomography [PET] scans) considering genetic and demographic factors as possible modifiers of these markers' progression. RESULTS Aβ42/40 ratio concentrations decreased, while NfL and GFAP values increased over the 4-year follow-up. Apolipoprotein E (APOE) ε4 carriers showed faster increase in plasma pTau181 than non-carriers. Older individuals showed a faster increase in plasma NfL, and females showed a faster increase in plasma GFAP values. In the PET subsample, individuals both Aβ-PET and tau-PET positive showed faster plasma pTau181 and GFAP increase compared to PET-negative individuals. DISCUSSION Plasma markers can track biological change over time, with plasma pTau181 and GFAP markers showing longitudinal change in individuals with preclinical AD. HIGHLIGHTS Longitudinal increase of plasma pTau181 and glial fibrillary acidic protein (GFAP) can be measured in the preclinical phase of AD. Apolipoprotein E ε4 carriers experience faster increase in plasma pTau181 over time than non-carriers. Female sex showed accelerated increase in plasma GFAP over time compared to males. Aβ42/40 and pTau231 values are already abnormal at baseline in individuals with both amyloid and tau PET burden.
Collapse
Affiliation(s)
- Yara Yakoub
- Douglas Mental Health University Institute, Centre for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Cherie Strikwerda-Brown
- Douglas Mental Health University Institute, Centre for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada
| | - Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Przemysław R Kac
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fernando Gonzalez-Ortiz
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jonathan Gallego-Rudolf
- Douglas Mental Health University Institute, Centre for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada
| | - Pierre-François Meyer
- Douglas Mental Health University Institute, Centre for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada
| | - Frédéric St-Onge
- Douglas Mental Health University Institute, Centre for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Jean-Paul Soucy
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - John C S Breitner
- Douglas Mental Health University Institute, Centre for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
- McGill Centre for Integrative Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- UW Department of Medicine, School of Medicine and Public Health, Madison, WI, USA
| | - 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
| | - Judes Poirier
- Douglas Mental Health University Institute, Centre for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Sylvia Villeneuve
- Douglas Mental Health University Institute, Centre for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
25
|
Burnham SC, Iaccarino L, Pontecorvo MJ, Fleisher AS, Lu M, Collins EC, Devous MD. A review of the flortaucipir literature for positron emission tomography imaging of tau neurofibrillary tangles. Brain Commun 2023; 6:fcad305. [PMID: 38187878 PMCID: PMC10768888 DOI: 10.1093/braincomms/fcad305] [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: 05/04/2023] [Revised: 09/13/2023] [Accepted: 11/14/2023] [Indexed: 01/09/2024] Open
Abstract
Alzheimer's disease is defined by the presence of β-amyloid plaques and neurofibrillary tau tangles potentially preceding clinical symptoms by many years. Previously only detectable post-mortem, these pathological hallmarks are now identifiable using biomarkers, permitting an in vivo definitive diagnosis of Alzheimer's disease. 18F-flortaucipir (previously known as 18F-T807; 18F-AV-1451) was the first tau positron emission tomography tracer to be introduced and is the only Food and Drug Administration-approved tau positron emission tomography tracer (Tauvid™). It has been widely adopted and validated in a number of independent research and clinical settings. In this review, we present an overview of the published literature on flortaucipir for positron emission tomography imaging of neurofibrillary tau tangles. We considered all accessible peer-reviewed literature pertaining to flortaucipir through 30 April 2022. We found 474 relevant peer-reviewed publications, which were organized into the following categories based on their primary focus: typical Alzheimer's disease, mild cognitive impairment and pre-symptomatic populations; atypical Alzheimer's disease; non-Alzheimer's disease neurodegenerative conditions; head-to-head comparisons with other Tau positron emission tomography tracers; and technical considerations. The available flortaucipir literature provides substantial evidence for the use of this positron emission tomography tracer in assessing neurofibrillary tau tangles in Alzheimer's disease and limited support for its use in other neurodegenerative disorders. Visual interpretation and quantitation approaches, although heterogeneous, mostly converge and demonstrate the high diagnostic and prognostic value of flortaucipir in Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | - Ming Lu
- Avid, Eli Lilly and Company, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
26
|
Kwon HS, Lee E, Kim H, Park S, Park H, Jeong JH, Koh S, Choi SH, Lee J. Predicting amyloid PET positivity using plasma p-tau181 and other blood-based biomarkers. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12502. [PMID: 38026758 PMCID: PMC10654468 DOI: 10.1002/dad2.12502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/12/2023] [Accepted: 10/22/2023] [Indexed: 12/01/2023]
Abstract
Introduction This study aimed to determine the efficacy of combining plasma phosphorylated tau (p-tau)181, amyloid beta (Aβ)42/Aβ40, neurofilament light (NfL), and apolipoprotein E (APOE) genotypes for detecting positive amyloid positron emission tomography (PET), which is little known in the Asian population, in two independent cohorts. Methods Biomarkers were measured using a single-molecule array (Simoa) in a cohort study (Asan). All participants underwent amyloid PET. Significant changes in the area under the curve (AUC) and Akaike Information Criterion values were considered to determine the best model. The generalizability of this model was tested using another cohort (KBASE-V). Results In the Asan cohort, after adjusting for age and sex, p-tau181 (AUC = 0.854) or APOE ε4 status (AUC = 0.769) distinguished Aβ status with high accuracy. Combining them or adding NfL and Aβ42/40 improved model fitness. The best-fit model included the plasma p-tau181, APOE ε4, NfL and Aβ42/40. The models established from the Asan cohort were tested in the KBASE-V cohort. Additionally, in the KBASE-V cohort, these three biomarker models had similar AUC in cognitively unimpaired (AUC = 0.768) and mild cognitive impairment (MCI) (AUC = 0.997) participants. Conclusions Plasma p-tau181 showed a high performance in determining Aβ-PET positivity. Adding plasma NfL and APOE ε4 status improved the model fit without significant improvement in AUC.
Collapse
Affiliation(s)
- Hyuk Sung Kwon
- Department of NeurologyHanyang University Guri HospitalHanyang University College of MedicineGuriSouth Korea
| | - Eun‐Hye Lee
- Department of NeurologyHanyang University Guri HospitalHanyang University College of MedicineGuriSouth Korea
| | - Hyung‐Ji Kim
- Department of NeurologyUijeongbu Eulji Medical CenterEulji UniversityUijeongbuSouth Korea
| | - So‐Hee Park
- Department of NeurologyBobath Memorial HospitalSeongnamSouth Korea
| | - Hyun‐Hee Park
- Department of NeurologyHanyang University Guri HospitalHanyang University College of MedicineGuriSouth Korea
| | - Jee Hyang Jeong
- Department of NeurologyEwha Womans University School of MedicineSeoulSouth Korea
| | - Seong‐Ho Koh
- Department of NeurologyHanyang University Guri HospitalHanyang University College of MedicineGuriSouth Korea
- Department of Translational MedicineHanyang University Graduate School of Biomedical Science & EngineeringSeoulSouth Korea
| | - Seong Hye Choi
- Department of NeurologyInha University College of MedicineIncheonSouth Korea
| | - Jae‐Hong Lee
- Department of NeurologyUniversity of Ulsan College of MedicineAsan Medical CenterSeoulSouth Korea
| |
Collapse
|
27
|
Ferreira PCL, Zhang Y, Snitz B, Chang CCH, Bellaver B, Jacobsen E, Kamboh MI, Zetterberg H, Blennow K, Pascoal TA, Villemagne VL, Ganguli M, Karikari TK. Plasma biomarkers identify older adults at risk of Alzheimer's disease and related dementias in a real-world population-based cohort. Alzheimers Dement 2023; 19:4507-4519. [PMID: 36876954 PMCID: PMC10480336 DOI: 10.1002/alz.12986] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/12/2023] [Accepted: 01/15/2023] [Indexed: 03/07/2023]
Abstract
INTRODUCTION Plasma biomarkers-cost effective, non-invasive indicators of Alzheimer's disease (AD) and related disorders (ADRD)-have largely been studied in clinical research settings. Here, we examined plasma biomarker profiles and their associated factors in a population-based cohort to determine whether they could identify an at-risk group, independently of brain and cerebrospinal fluid biomarkers. METHODS We measured plasma phosphorylated tau181 (p-tau181), neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), and amyloid beta (Aβ)42/40 ratio in 847 participants from a population-based cohort in southwestern Pennsylvania. RESULTS K-medoids clustering identified two distinct plasma Aβ42/40 modes, further categorizable into three biomarker profile groups: normal, uncertain, and abnormal. In different groups, plasma p-tau181, NfL, and GFAP were inversely correlated with Aβ42/40, Clinical Dementia Rating, and memory composite score, with the strongest associations in the abnormal group. DISCUSSION Abnormal plasma Aβ42/40 ratio identified older adult groups with lower memory scores, higher dementia risks, and higher ADRD biomarker levels, with potential implications for population screening. HIGHLIGHTS Population-based plasma biomarker studies are lacking, particularly in cohorts without cerebrospinal fluid or neuroimaging data. In the Monongahela-Youghiogheny Healthy Aging Team study (n = 847), plasma biomarkers associated with worse memory and Clinical Dementia Rating (CDR), apolipoprotein E ε4, and greater age. Plasma amyloid beta (Aβ)42/40 ratio levels allowed clustering participants into abnormal, uncertain, and normal groups. Plasma Aβ42/40 correlated differently with neurofilament light chain, glial fibrillary acidic protein, phosphorylated tau181, memory composite, and CDR in each group. Plasma biomarkers can enable relatively affordable and non-invasive community screening for evidence of Alzheimer's disease and related disorders pathophysiology.
Collapse
Affiliation(s)
- Pamela C. L Ferreira
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Yingjin Zhang
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Beth Snitz
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Chung-Chou H. Chang
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Bruna Bellaver
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Erin Jacobsen
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - M. Ilyas Kamboh
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1N 3BG, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, HKG, China
- UW Department of Medicine, School of Medicine and Public Health, Madison, WI, 53726, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
| | - Tharick A. Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Victor L. Villemagne
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Mary Ganguli
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Thomas K. Karikari
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Mölndal, 431 41, Sweden
| |
Collapse
|
28
|
Bilgel M, An Y, Walker KA, Moghekar AR, Ashton NJ, Kac PR, Karikari TK, Blennow K, Zetterberg H, Jedynak BM, Thambisetty M, Ferrucci L, Resnick SM. Longitudinal changes in Alzheimer's-related plasma biomarkers and brain amyloid. Alzheimers Dement 2023; 19:4335-4345. [PMID: 37216632 PMCID: PMC10592628 DOI: 10.1002/alz.13157] [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: 02/08/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/24/2023]
Abstract
INTRODUCTION Understanding longitudinal plasma biomarker trajectories relative to brain amyloid changes can help devise Alzheimer's progression assessment strategies. METHODS We examined the temporal order of changes in plasma amyloid-β ratio (A β 42 / A β 40 ${{\rm A}\beta }_{42}/{{\rm A}\beta }_{40}$ ), glial fibrillary acidic protein (GFAP), neurofilament light chain (NfL), and phosphorylated tau ratios (p-tau181 / A β 42 $\text{p-tau181}/\mathrm{A}{\beta}_{42}$ ,p-tau231 / A β 42 $\text{p-tau231}/\mathrm{A}{\beta}_{42}$ ) relative to 11 C-Pittsburgh compound B (PiB) positron emission tomography (PET) cortical amyloid burden (PiB-/+). Participants (n = 199) were cognitively normal at index visit with a median 6.1-year follow-up. RESULTS PiB groups exhibited different rates of longitudinal change inA β 42 / A β 40 ( β = 5.41 × 10 - 4 , SE = 1.95 × 10 - 4 , p = 0.0073 ) ${{\rm A}\beta }_{42}/{{\rm A}\beta }_{40}\ ( {\beta \ = \ 5.41 \times {{10}}^{ - 4},{\rm{\ SE\ }} = \ 1.95 \times {{10}}^{ - 4},\ p\ = \ 0.0073} )$ . Change in brain amyloid correlated with change in GFAP (r = 0.5, 95% CI = [0.26, 0.68]). The greatest relative decline inA β 42 / A β 40 ${{\rm A}\beta }_{42}/{{\rm A}\beta }_{40}$ (-1%/year) preceded brain amyloid positivity by 41 years (95% CI = [32, 53]). DISCUSSION PlasmaA β 42 / A β 40 ${{\rm A}\beta }_{42}/{{\rm A}\beta }_{40}$ may begin declining decades prior to brain amyloid accumulation, whereas p-tau ratios, GFAP, and NfL increase closer in time. HIGHLIGHTS PlasmaA β 42 / A β 40 ${{\rm A}\beta }_{42}/{{\rm A}\beta }_{40}$ declines over time among PiB- but does not change among PiB+. Phosphorylated-tau to Aβ42 ratios increase over time among PiB+ but do not change among PiB-. Rate of change in brain amyloid is correlated with change in GFAP and neurofilament light chain. The greatest decline inA β 42 / A β 40 ${{\rm A}\beta }_{42}/{{\rm A}\beta }_{40}$ may precede brain amyloid positivity by decades.
Collapse
Affiliation(s)
- Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, 21224, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, 21224, USA
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, 21224, USA
| | - Abhay R. Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 80 Mölndal, Sweden
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl 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
- Centre for Age-Related Medicine, Stavanger University Hospital, 4019 Stavanger, Norway
| | - Przemysław R. Kac
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 80 Mölndal, Sweden
| | - Thomas K. Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 80 Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 80 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 80 Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 80 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 80 Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, 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
| | - Bruno M. Jedynak
- Department of Mathematics and Statistics, Portland State University, Portland, Oregon, 97201, USA
| | - Madhav Thambisetty
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, 21224, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland, 21224, USA
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, 21224, USA
| |
Collapse
|
29
|
Pais MV, Kuo C, Ances BM, Wetherell JL, Lenze EJ, Diniz BS. Relationship between baseline plasma p-tau181 and longitudinal changes in cognition and structural brain measures in a cohort of cognitively unimpaired older adults. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12487. [PMID: 37954547 PMCID: PMC10634375 DOI: 10.1002/dad2.12487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/22/2023] [Indexed: 11/14/2023]
Abstract
INTRODUCTION Preclinical Alzheimer's disease (AD) affects a significant proportion of cognitively unimpaired (CU) older adults. Currently, blood-based biomarkers detect very early changes in the AD continuum with great accuracy. METHODS We measured baseline plasma phosphorylated tau (p-tau)181 using electrochemiluminescence (ECL)-based assay (MesoScale Discovery) in 533 CU older adults. Follow-up lasted up to 18 months. Cognitive performance assessment included memory and cognitive control. Structural brain measures included cortical thickness, which includes the AD magnetic resonance imaging (AD MRI) signature, and hippocampal volume. RESULTS In this cohort of CU older adults, baseline plasma p-tau181 levels were not associated with short-term changes in cognition and structural brain measures. Also, baseline plasma p-tau levels did not influence the effects of behavioral interventions (exercise or mindfulness) on cognitive and structural brain changes. DISCUSSION The short follow-up and healthy status of this CU cohort might have limited the sensitivity of plasma p-tau181 in detecting changes associated with AD pathology.
Collapse
Affiliation(s)
- Marcos V. Pais
- UConn Center on AgingUniversity of Connecticut Health CenterFarmingtonConnecticutUSA
- Laboratory of Neuroscience (LIM‐27)Departamento e Instituto de PsiquiatriaFaculdade de Medicina, Universidade de Sao Paulo (FMUSP)Sao PauloBrazil
| | - Chia‐Ling Kuo
- Department of Public Health SciencesUniversity of Connecticut Health CenterFarmingtonConnecticutUSA
| | - Beau M. Ances
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | | | - Eric J. Lenze
- Healthy Mind Lab, Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
| | - Breno S. Diniz
- UConn Center on AgingUniversity of Connecticut Health CenterFarmingtonConnecticutUSA
| |
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
|
Estepp TG, Charnigo RJ, Abner EL, Jicha GA, Sudduth TL, Fardo DW, Wilcock DM. Associations of potential ADRD plasma biomarkers in cognitively normal volunteers. Alzheimers Dement 2023; 19:3593-3601. [PMID: 36840666 PMCID: PMC10440211 DOI: 10.1002/alz.13000] [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: 08/29/2022] [Revised: 01/18/2023] [Accepted: 01/22/2023] [Indexed: 02/26/2023]
Abstract
INTRODUCTION This study examined the relationships between 13 novel blood-plasma biomarkers and dementia-related demographic and health factors in a cohort of 237 cognitively normal research volunteers whose average age was ≈82 years and who were 63% female. METHODS We regressed each biomarker on selected covariates to explore the associations between the biomarkers and selected factors to assess whether they may contribute to biomarker values. Post hoc sensitivity analyses were done with updated data and consistent variable sets for robustness and batch effects. RESULTS Biomarker concentrations were largely not associated with demographics or health conditions, but some expected associations (e.g., apolipoprotein E [APOE] status with amyloid beta [Aβ]42/Aβ40) were observed. Post hoc results remained similar to those of the main analysis. DISCUSSION The absence of strong associations between the biomarkers with age, gender, or medical conditions suggests that changes in these biomarkers, when observed, may be attributable to neuropathological changes. HIGHLIGHTS Among N = 237 cognitively normal adults, we studied candidate Alzheimer's disease and related dementia (ADRD) plasma biomarkers. Biomarkers were largely not associated with demographic or health factors. Apolipoprotein E (APOE) status was associated with amyloid beta (Aβ)42/Aβ40 ratio. These results support hypotheses that plasma biomarkers are informative for ADRD.
Collapse
Affiliation(s)
- Taylor G. Estepp
- Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky, 800 S. Limestone St, Lexington, KY, 40536, USA
- College of Public Health, Department of Epidemiology, University of Kentucky, 111 Washington Ave, Lexington, KY, 40536, USA
- College of Public Health, Department of Biostatistics, University of Kentucky, 111 Washington Ave, Lexington, KY, 40536, USA
| | - Richard J. Charnigo
- College of Public Health, Department of Biostatistics, University of Kentucky, 111 Washington Ave, Lexington, KY, 40536, USA
- College of Arts and Sciences, Department of Statistics, University of Kentucky, 725 Rose St, Lexington, KY, 40536, USA
| | - Erin L. Abner
- Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky, 800 S. Limestone St, Lexington, KY, 40536, USA
- College of Public Health, Department of Epidemiology, University of Kentucky, 111 Washington Ave, Lexington, KY, 40536, USA
- College of Public Health, Department of Biostatistics, University of Kentucky, 111 Washington Ave, Lexington, KY, 40536, USA
| | - Gregory A. Jicha
- Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky, 800 S. Limestone St, Lexington, KY, 40536, USA
- College of Medicine, Department of Neurology, University of Kentucky, 740 S. Limestone, Lexington, KY, 40536, USA
| | - Tiffany L. Sudduth
- Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky, 800 S. Limestone St, Lexington, KY, 40536, USA
| | - David W. Fardo
- Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky, 800 S. Limestone St, Lexington, KY, 40536, USA
- College of Public Health, Department of Biostatistics, University of Kentucky, 111 Washington Ave, Lexington, KY, 40536, USA
| | - Donna M. Wilcock
- Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky, 800 S. Limestone St, Lexington, KY, 40536, USA
- College of Medicine, Department of Physiology, University of Kentucky, 780 Rose St, Lexington, KY, 40536, USA
| |
Collapse
|
32
|
Xu C, Zhao L, Dong C. The performance of plasma phosphorylated tau231 in detecting Alzheimer's disease: A systematic review with meta-analysis. Eur J Neurosci 2023; 58:3132-3149. [PMID: 37501373 DOI: 10.1111/ejn.16085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/15/2023] [Accepted: 06/26/2023] [Indexed: 07/29/2023]
Abstract
Cerebrospinal fluid (CSF) phosphorylated tau231 (P-tau231) is associated with neuropathological outcomes of Alzheimer's disease (AD). The invasive access of cerebrospinal fluid has greatly stimulated interest in the identification of blood-based P-tau231, and the recent advent of single-molecule array assay for the quantification of plasma P-tau231 may provide a turning point to evaluate the usefulness of P-tau231 as an AD-related biomarker. Yet, in the plasma P-tau231 literature, findings with regard to its diagnostic utility have been inconsistent, and thus, we aimed to statistically investigate the potential of plasma P-tau231 in the context of AD via meta-analysis. Publications on plasma P-tau231 were systematically retrieved from PubMed, EMBASE, the Cochrane library and Web of Science databases. A total of 10 studies covering 2007 participants were included, and we conducted random-effect or fixed-effect meta-analysis, sensitivity analysis and publication bias analysis using the STATA SE 14.0 software. According to our quantitative integration, plasma P-tau231 increased from cognitively unimpaired (CU) populations to mild cognitive impairment to AD and showed significant changes in pairwise comparisons of AD, mild cognitive impairment and CU. Plasma P-tau231 level was significantly higher in CU controls with positive amyloid-β (Aβ) status compared with Aβ-negative CU group. Additionally, the excellent diagnostic accuracy of plasma P-tau231 for asymptomatic Aβ pathology was verified by the pooled value of area under the receiver operating characteristic curves (standard mean difference [95% confidence interval]: .75 [.69, .81], P < 0.00001). Overall, the increased plasma P-tau231 concentrations were found in relation to the early development and progression of AD.
Collapse
Affiliation(s)
- Chang Xu
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Li Zhao
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Chunbo Dong
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
33
|
Snellman A, Ekblad LL, Ashton NJ, Karikari TK, Lantero-Rodriguez J, Pietilä E, Koivumäki M, Helin S, Karrasch M, Zetterberg H, Blennow K, Rinne JO. Head-to-head comparison of plasma p-tau181, p-tau231 and glial fibrillary acidic protein in clinically unimpaired elderly with three levels of APOE4-related risk for Alzheimer's disease. Neurobiol Dis 2023; 183:106175. [PMID: 37268240 DOI: 10.1016/j.nbd.2023.106175] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 05/02/2023] [Accepted: 05/26/2023] [Indexed: 06/04/2023] Open
Abstract
Plasma phosphorylated tau (p-tau) and glial fibrillary acidic protein (GFAP) both reflect early changes in Alzheimer's disease (AD) pathology. Here, we compared the biomarker levels and their association with regional β-amyloid (Aβ) pathology and cognitive performance head-to-head in clinically unimpaired elderly (n = 88) at three levels of APOE4-related genetic risk for sporadic AD (APOE4/4 n = 19, APOE3/4 n = 32 or non-carriers n = 37). Concentrations of plasma p-tau181, p-tau231 and GFAP were measured using Single molecule array (Simoa), regional Aβ deposition with 11C-PiB positron emission tomography (PET), and cognitive performance with a preclinical composite. Significant differences in plasma p-tau181 and p-tau231, but not plasma GFAP concentrations were present between the APOE4 gene doses, explained solely by brain Aβ load. All plasma biomarkers correlated positively with Aβ PET in the total study population. This correlation was driven by APOE3/3 carriers for plasma p-tau markers and APOE4/4 carriers for plasma GFAP. Voxel-wise associations with amyloid-PET revealed different spatial patterns for plasma p-tau markers and plasma GFAP. Only higher plasma GFAP correlated with lower cognitive scores. Our observations suggest that plasma p-tau and plasma GFAP are both early AD markers reflecting different Aβ-related processes.
Collapse
Affiliation(s)
- Anniina Snellman
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland; Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
| | - Laura L Ekblad
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway; Department of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Elina Pietilä
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
| | - Mikko Koivumäki
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
| | - Semi Helin
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
| | - Mira Karrasch
- Department of Psychology, Åbo Akademi University, Turku, Finland
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; UK Dementia Research Institute at UCL, London, UK; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, 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, WI 53792, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Juha O Rinne
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland; InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| |
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
|
Luebke M, Parulekar M, Thomas FP. Fluid biomarkers for the diagnosis of neurodegenerative diseases. Biomark Neuropsychiatry 2023. [DOI: 10.1016/j.bionps.2023.100062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
|
36
|
Van Egroo M, Riphagen JM, Ashton NJ, Janelidze S, Sperling RA, Johnson KA, Yang HS, Bennett DA, Blennow K, Hansson O, Zetterberg H, Jacobs HIL. Ultra-high field imaging, plasma markers and autopsy data uncover a specific rostral locus coeruleus vulnerability to hyperphosphorylated tau. Mol Psychiatry 2023; 28:2412-2422. [PMID: 37020050 PMCID: PMC10073793 DOI: 10.1038/s41380-023-02041-y] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 04/07/2023]
Abstract
Autopsy data indicate that the locus coeruleus (LC) is one of the first sites in the brain to accumulate hyperphosphorylated tau pathology, with the rostral part possibly being more vulnerable in the earlier stages of the disease. Taking advantage of recent developments in ultra-high field (7 T) imaging, we investigated whether imaging measures of the LC also reveal a specific anatomic correlation with tau using novel plasma biomarkers of different species of hyperphosphorylated tau, how early in adulthood these associations can be detected and if are associated with worse cognitive performance. To validate the anatomic correlations, we tested if a rostro-caudal gradient in tau pathology is also detected at autopsy in data from the Rush Memory and Aging Project (MAP). We found that higher plasma measures of phosphorylated tau, in particular ptau231, correlated negatively with dorso-rostral LC integrity, whereas correlations for neurodegenerative plasma markers (neurofilament light, total tau) were scattered throughout the LC including middle to caudal sections. In contrast, the plasma Aβ42/40 ratio, associated with brain amyloidosis, did not correlate with LC integrity. These findings were specific to the rostral LC and not observed when using the entire LC or the hippocampus. Furthermore, in the MAP data, we observed higher rostral than caudal tangle density in the LC, independent of the disease stage. The in vivo LC-phosphorylated tau correlations became significant from midlife, with the earliest effect for ptau231, starting at about age 55. Finally, interactions between lower rostral LC integrity and higher ptau231 concentrations predicted lower cognitive performance. Together, these findings demonstrate a specific rostral vulnerability to early phosphorylated tau species that can be detected with dedicated magnetic resonance imaging measures, highlighting the promise of LC imaging as an early marker of AD-related processes.
Collapse
Grants
- R01 AG017917 NIA NIH HHS
- R01 AG068398 NIA NIH HHS
- R21 AG074220 NIA NIH HHS
- K23 AG062750 NIA NIH HHS
- R01 AG068062 NIA NIH HHS
- K01 AG001016 NIA NIH HHS
- ZEN-21-848495 Alzheimer's Association
- P01 AG036694 NIA NIH HHS
- R01 AG062559 NIA NIH HHS
- R01 AG015819 NIA NIH HHS
- U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- Alzheimer Nederland WE.03-2019-02
- BrightFocus Foundation (BrightFocus)
- Alzheimer’s Association
- Alzheimer’s Drug Discovery Foundation (ADDF)
- Swedish Research Council (#2017-00915), the Alzheimer Drug Discovery Foundation (ADDF), USA (#RDAPB-201809-2016615), the Swedish Alzheimer Foundation (#AF-930351, #AF-939721 and #AF-968270), Hjärnfonden, Sweden (#FO2017-0243 and #ALZ2022-0006), the Swedish state under the agreement between the Swedish government and the County Councils, the ALF-agreement (#ALFGBG-715986 and #ALFGBG-965240), the European Union Joint Program for Neurodegenerative Disorders (JPND2019-466-236)
- Cure Alzheimer’s Fund (Alzheimer’s Disease Research Foundation)
- Swedish Research Council (2016-00906), the Knut and Alice Wallenberg foundation (2017-0383), the Marianne and Marcus Wallenberg foundation (2015.0125), the Strategic Research Area MultiPark (Multidisciplinary Research in Parkinson’s disease) at Lund University, the Swedish Alzheimer Foundation (AF-939932), the Swedish Brain Foundation (FO2021-0293), The Parkinson foundation of Sweden (1280/20), the Cure Alzheimer’s fund, the Konung Gustaf V:s och Drottning Victorias Frimurarestiftelse, the Skåne University Hospital Foundation (2020-O000028), Regionalt Forskningsstöd (2020-0314) and the Swedish federal government under the ALF agreement (2018-Projekt0279)
- HZ is a Wallenberg Scholar supported by grants from the Swedish Research Council (#2018-02532), the European Research Council (#681712 and #101053962), Swedish State Support for Clinical Research (#ALFGBG-71320), the Alzheimer Drug Discovery Foundation (ADDF), USA (#201809-2016862), the AD Strategic Fund and the Alzheimer’s Association (#ADSF-21-831376-C, #ADSF-21-831381-C, and #ADSF-21-831377-C), the Bluefield Project, the Olav Thon Foundation, the Erling-Persson Family Foundation, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden (#FO2022-0270), the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement No 860197 (MIRIADE), the European Union Joint Programme – Neurodegenerative Disease Research (JPND2021-00694), and the UK Dementia Research Institute at UCL (UKDRI-1003).
Collapse
Affiliation(s)
- Maxime Van Egroo
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Joost M Riphagen
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Reisa A Sperling
- Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Keith A Johnson
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Hyun-Sik Yang
- Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - David A Bennett
- Department of Neurological Sciences, Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- 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, Clear Water Bay, Hong Kong, China
| | - Heidi I L Jacobs
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands.
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
37
|
Jiang X, Cui L, Huang L, Guo Y, Huang G, Guo Q. The Relationship between Beverages Consumption and Cognitive Impairment in Middle-Aged and Elderly Chinese Population. Nutrients 2023; 15:nu15102309. [PMID: 37242194 DOI: 10.3390/nu15102309] [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: 03/18/2023] [Revised: 04/23/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Some evidence shows that beverage consumption has an impact on cognitive performance. This is a follow-up study of dietary habits and cognitive function in the Chinese middle-aged and elderly population. The objective of this study was to explore the relationship between beverage consumption and cognitive impairment. The source and grouping of the participants can be seen in the previous article, "Study of Diet Habits and Cognitive Function in the Chinese Middle-Aged and Elderly Population: The Association between Folic Acid, B Vitamins, Vitamin D, Coenzyme Q10 Supplementation and Cognitive Ability". Among 892 participants, one-third (296) completed both Amyloid beta(Aβ)-PET and plasma biomarkers. The results showed that the consumption of beverages (green tea, coffee, pure milk) was a protective factor for cognitive impairment, daily water consumption <1500 mL (especially <500 mL) was a risk factor for cognitive impairment, and the above correlated with baseline cognitive status. The relationship of green tea, coffee, and pure milk consumption with cognitive impairment was related to gender. We also found that among the participants with Aβ deposition, the consumption of pure milk and green tea was associated with low levels of p-Tau-181. In conclusion, the relationship between beverage consumption and cognitive impairment in Chinese middle-aged and elderly adults may be related to baseline cognitive status, gender, and Aβ deposition.
Collapse
Affiliation(s)
- Xinting Jiang
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Department of VIP Clinical, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Liang Cui
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Lin Huang
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yihan Guo
- Faculty of Medicine, The University of Queensland, Brisbane 4072, Australia
| | - Gaozhong Huang
- Department of VIP Clinical, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Qihao Guo
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| |
Collapse
|
38
|
Jack CR, Wiste HJ, Algeciras-Schimnich A, Figdore DJ, Schwarz CG, Lowe VJ, Ramanan VK, Vemuri P, Mielke MM, Knopman DS, Graff-Radford J, Boeve BF, Kantarci K, Cogswell PM, Senjem ML, Gunter JL, Therneau TM, Petersen RC. Predicting amyloid PET and tau PET stages with plasma biomarkers. Brain 2023; 146:2029-2044. [PMID: 36789483 PMCID: PMC10151195 DOI: 10.1093/brain/awad042] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/20/2022] [Accepted: 01/21/2023] [Indexed: 02/16/2023] Open
Abstract
Staging the severity of Alzheimer's disease pathology using biomarkers is useful for therapeutic trials and clinical prognosis. Disease staging with amyloid and tau PET has face validity; however, this would be more practical with plasma biomarkers. Our objectives were, first, to examine approaches for staging amyloid and tau PET and, second, to examine prediction of amyloid and tau PET stages using plasma biomarkers. Participants (n = 1136) were enrolled in either the Mayo Clinic Study of Aging or the Alzheimer's Disease Research Center; had a concurrent amyloid PET, tau PET and blood draw; and met clinical criteria for cognitively unimpaired (n = 864), mild cognitive impairment (n = 148) or Alzheimer's clinical syndrome with dementia (n = 124). The latter two groups were combined into a cognitively impaired group (n = 272). We used multinomial regression models to estimate discrimination [concordance (C) statistics] among three amyloid PET stages (low, intermediate, high), four tau PET stages (Braak 0, 1-2, 3-4, 5-6) and a combined amyloid and tau PET stage (none/low versus intermediate/high severity) using plasma biomarkers as predictors separately within unimpaired and impaired individuals. Plasma analytes, p-tau181, Aβ1-42 and Aβ1-40 (analysed as the Aβ42/Aβ40 ratio), glial fibrillary acidic protein and neurofilament light chain were measured on the HD-X Simoa Quanterix platform. Plasma p-tau217 was also measured in a subset (n = 355) of cognitively unimpaired participants using the Lilly Meso Scale Discovery assay. Models with all Quanterix plasma analytes along with risk factors (age, sex and APOE) most often provided the best discrimination among amyloid PET stages (C = 0.78-0.82). Models with p-tau181 provided similar discrimination of tau PET stages to models with all four plasma analytes (C = 0.72-0.85 versus C = 0.73-0.86). Discriminating a PET proxy of intermediate/high from none/low Alzheimer's disease neuropathological change with all four Quanterix plasma analytes was excellent but not better than p-tau181 only (C = 0.88 versus 0.87 for unimpaired and C = 0.91 versus 0.90 for impaired). Lilly p-tau217 outperformed the Quanterix p-tau181 assay for discriminating high versus intermediate amyloid (C = 0.85 versus 0.74) but did not improve over a model with all Quanterix plasma analytes and risk factors (C = 0.85 versus 0.83). Plasma analytes along with risk factors can discriminate between amyloid and tau PET stages and between a PET surrogate for intermediate/high versus none/low neuropathological change with accuracy in the acceptable to excellent range. Combinations of plasma analytes are better than single analytes for many staging predictions with the exception that Quanterix p-tau181 alone usually performed equivalently to combinations of Quanterix analytes for tau PET discrimination.
Collapse
Affiliation(s)
- Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Heather J Wiste
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Dan J Figdore
- Department of Laboratory Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Val J Lowe
- Department of Nuclear Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Vijay K Ramanan
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Michelle M Mielke
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Bradley F Boeve
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | - Terry M Therneau
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
39
|
Bilgel M, An Y, Walker KA, Moghekar AR, Ashton NJ, Kac PR, Karikari TK, Blennow K, Zetterberg H, Jedynak BM, Thambisetty M, Ferrucci L, Resnick SM. Longitudinal changes in Alzheimer's-related plasma biomarkers and brain amyloid. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.01.12.23284439. [PMID: 36711545 PMCID: PMC9882432 DOI: 10.1101/2023.01.12.23284439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Understanding longitudinal plasma biomarker trajectories relative to brain amyloid changes can help devise Alzheimer's progression assessment strategies. METHODS We examined the temporal order of changes in plasma amyloid-β ratio (Aβ 42 /Aβ 40 ), glial fibrillary acidic protein (GFAP), neurofilament light chain (NfL), and phosphorylated tau ratios (p-tau181/Aβ 42 , p-tau231/Aβ 42 ) relative to 11 C-Pittsburgh compound B (PiB) positron emission tomography (PET) cortical amyloid burden (PiB-/+). Participants (n = 199) were cognitively normal at index visit with a median 6.1-year follow-up. RESULTS PiB groups exhibited different rates of longitudinal change in Aβ 42 /Aβ 40 (β = 5.41 × 10^ -4 , SE = 1.95 × 10 -4 , p = 0.0073). Change in brain amyloid was correlated with change in GFAP (r = 0.5, 95% CI = [0.26, 0.68]). Greatest relative decline in Aβ 42 /Aβ 40 (-1%/year) preceded brain amyloid positivity onset by 41 years (95% CI = [32, 53]). DISCUSSION Plasma Aβ 42 /Aβ 40 may begin declining decades prior to brain amyloid accumulation, whereas p-tau ratios, GFAP, and NfL increase closer in time.
Collapse
Affiliation(s)
- Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, 21224, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, 21224, USA
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, 21224, USA
| | - Abhay R. Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 80 Mölndal, Sweden
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl 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
- Centre for Age-Related Medicine, Stavanger University Hospital, 4019 Stavanger, Norway
| | - Przemyslaw R. Kac
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 80 Mölndal, Sweden
| | - Thomas K. Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 80 Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 80 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 80 Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 80 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 80 Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, 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
| | - Bruno M. Jedynak
- Department of Mathematics and Statistics, Portland State University, Portland, Oregon, 97201, USA
| | - Madhav Thambisetty
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, 21224, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland, 21224, USA
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, 21224, USA
| |
Collapse
|
40
|
Saunders TS, Pozzolo FE, Heslegrave A, King D, McGeachan RI, Spires-Jones MP, Harris SE, Ritchie C, Muniz-Terrera G, Deary IJ, Cox SR, Zetterberg H, Spires-Jones TL. Predictive blood biomarkers and brain changes associated with age-related cognitive decline. Brain Commun 2023; 5:fcad113. [PMID: 37180996 PMCID: PMC10167767 DOI: 10.1093/braincomms/fcad113] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 12/28/2022] [Accepted: 04/05/2023] [Indexed: 04/08/2023] Open
Abstract
Growing evidence supports the use of plasma levels of tau phosphorylated at threonine 181, amyloid-β, neurofilament light and glial fibrillary acidic protein as promising biomarkers for Alzheimer's disease. While these blood biomarkers are promising for distinguishing people with Alzheimer's disease from healthy controls, their predictive validity for age-related cognitive decline without dementia remains unclear. Further, while tau phosphorylated at threonine 181 is a promising biomarker, the distribution of this phospho-epitope of tau in the brain is unknown. Here, we tested whether plasma levels of tau phosphorylated at threonine 181, amyloid-β, neurofilament light and fibrillary acidic protein predict cognitive decline between ages 72 and 82 in 195 participants in the Lothian birth cohorts 1936 study of cognitive ageing. We further examined post-mortem brain samples from temporal cortex to determine the distribution of tau phosphorylated at threonine 181 in the brain. Several forms of tau phosphorylated at threonine 181 have been shown to contribute to synapse degeneration in Alzheimer's disease, which correlates closely with cognitive decline in this form of dementia, but to date, there have not been investigations of whether tau phosphorylated at threonine 181 is found in synapses in Alzheimer's disease or healthy ageing brain. It was also previously unclear whether tau phosphorylated at threonine 181 accumulated in dystrophic neurites around plaques, which could contribute to tau leakage to the periphery due to impaired membrane integrity in dystrophies. Brain homogenate and biochemically enriched synaptic fractions were examined with western blot to examine tau phosphorylated at threonine 181 levels between groups (n = 10-12 per group), and synaptic and astrocytic localization of tau phosphorylated at threonine 181 were examined using array tomography (n = 6-15 per group), and localization of tau phosphorylated at threonine 181 in plaque-associated dystrophic neurites with associated gliosis were examined with standard immunofluorescence (n = 8-9 per group). Elevated baseline plasma tau phosphorylated at threonine 181, neurofilament light and fibrillary acidic protein predicted steeper general cognitive decline during ageing. Further, increasing tau phosphorylated at threonine 181 over time predicted general cognitive decline in females only. Change in plasma tau phosphorylated at threonine 181 remained a significant predictor of g factor decline when taking into account Alzheimer's disease polygenic risk score, indicating that the increase of blood tau phosphorylated at threonine 181 in this cohort was not only due to incipient Alzheimer's disease. Tau phosphorylated at threonine 181 was observed in synapses and astrocytes in both healthy ageing and Alzheimer's disease brain. We observed that a significantly higher proportion of synapses contain tau phosphorylated at threonine 181 in Alzheimer's disease relative to aged controls. Aged controls with pre-morbid lifetime cognitive resilience had significantly more tau phosphorylated at threonine 181 in fibrillary acidic protein-positive astrocytes than those with pre-morbid lifetime cognitive decline. Further, tau phosphorylated at threonine 181 was found in dystrophic neurites around plaques and in some neurofibrillary tangles. The presence of tau phosphorylated at threonine 181 in plaque-associated dystrophies may be a source of leakage of tau out of neurons that eventually enters the blood. Together, these data indicate that plasma tau phosphorylated at threonine 181, neurofilament light and fibrillary acidic protein may be useful biomarkers of age-related cognitive decline, and that efficient clearance of tau phosphorylated at threonine 181 by astrocytes may promote cognitive resilience.
Collapse
Affiliation(s)
- Tyler S Saunders
- UK Dementia Research Institute and Centre for Discovery Brain Sciences at the University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Edinburgh Dementia Prevention & Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Francesca E Pozzolo
- UK Dementia Research Institute and Centre for Discovery Brain Sciences at the University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Amanda Heslegrave
- United Kingdom UK Dementia Research Institute at University College London, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Declan King
- UK Dementia Research Institute and Centre for Discovery Brain Sciences at the University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Robert I McGeachan
- UK Dementia Research Institute and Centre for Discovery Brain Sciences at the University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Maxwell P Spires-Jones
- UK Dementia Research Institute and Centre for Discovery Brain Sciences at the University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Sarah E Harris
- Lothian Birth Cohort studies, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9AD, UK
| | - Craig Ritchie
- Edinburgh Dementia Prevention & Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Graciela Muniz-Terrera
- Edinburgh Dementia Prevention & Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH4 2XU, UK
- Department of Social Medicine, Ohio University, Athens, Ohio 45701, USA
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago 3485, Chile
| | - Ian J Deary
- Lothian Birth Cohort studies, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9AD, UK
| | - Simon R Cox
- Lothian Birth Cohort studies, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9AD, UK
| | - Henrik Zetterberg
- United Kingdom UK Dementia Research Institute at University College London, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, S-431 80 Molndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80 Molndal, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Tara L Spires-Jones
- UK Dementia Research Institute and Centre for Discovery Brain Sciences at the University of Edinburgh, Edinburgh, EH8 9JZ, UK
| |
Collapse
|
41
|
Snellman A, Ekblad LL, Tuisku J, Koivumäki M, Ashton NJ, Lantero-Rodriguez J, Karikari TK, Helin S, Bucci M, Löyttyniemi E, Parkkola R, Karrasch M, Schöll M, Zetterberg H, Blennow K, Rinne JO. APOE ε4 gene dose effect on imaging and blood biomarkers of neuroinflammation and beta-amyloid in cognitively unimpaired elderly. Alzheimers Res Ther 2023; 15:71. [PMID: 37016464 PMCID: PMC10071691 DOI: 10.1186/s13195-023-01209-6] [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: 09/29/2022] [Accepted: 03/13/2023] [Indexed: 04/06/2023]
Abstract
BACKGROUND Neuroinflammation, characterized by increased reactivity of microglia and astrocytes in the brain, is known to be present at various stages of the Alzheimer's disease (AD) continuum. However, its presence and relationship with amyloid pathology in cognitively normal at-risk individuals is less clear. Here, we used positron emission tomography (PET) and blood biomarker measurements to examine differences in neuroinflammation and beta-amyloid (Aβ) and their association in cognitively unimpaired homozygotes, heterozygotes, or non-carriers of the APOE ε4 allele, the strongest genetic risk for sporadic AD. METHODS Sixty 60-75-year-old APOE ε4 homozygotes (n = 19), heterozygotes (n = 21), and non-carriers (n = 20) were recruited in collaboration with the local Auria biobank. The participants underwent 11C-PK11195 PET (targeting 18-kDa translocator protein, TSPO), 11C-PiB PET (targeting Aβ), brain MRI, and neuropsychological testing including a preclinical cognitive composite (APCC). 11C-PK11195 distribution volume ratios and 11C-PiB standardized uptake value ratios (SUVRs) were calculated for regions typical for early Aβ accumulation in AD. Blood samples were drawn for measuring plasma glial fibrillary acidic protein (GFAP) and plasma Aβ1-42/1.40. RESULTS In our cognitively unimpaired sample, cortical 11C-PiB-binding increased according to APOE ε4 gene dose (median composite SUVR 1.47 (range 1.38-1.66) in non-carriers, 1.55 (1.43-2.02) in heterozygotes, and 2.13 (1.61-2.83) in homozygotes, P = 0.002). In contrast, cortical composite 11C-PK11195-binding did not differ between the APOE ε4 gene doses (P = 0.27) or between Aβ-positive and Aβ-negative individuals (P = 0.81) and associated with higher Aβ burden only in APOE ε4 homozygotes (Rho = 0.47, P = 0.043). Plasma GFAP concentration correlated with cortical 11C-PiB (Rho = 0.35, P = 0.040), but not 11C-PK11195-binding (Rho = 0.13, P = 0.47) in Aβ-positive individuals. In the total cognitively unimpaired population, both higher composite 11C-PK11195-binding and plasma GFAP were associated with lower hippocampal volume, whereas elevated 11C-PiB-binding was associated with lower APCC scores. CONCLUSIONS Only Aβ burden measured by PET, but not markers of neuroinflammation, differed among cognitively unimpaired elderly with different APOE ε4 gene dose. However, APOE ε4 gene dose seemed to modulate the association between neuroinflammation and Aβ.
Collapse
Affiliation(s)
- Anniina Snellman
- Turku PET Centre, University of Turku, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland.
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
| | - Laura L Ekblad
- Turku PET Centre, University of Turku, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Jouni Tuisku
- Turku PET Centre, University of Turku, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Mikko Koivumäki
- Turku PET Centre, University of Turku, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Department of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Semi Helin
- Turku PET Centre, University of Turku, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Marco Bucci
- Turku PET Centre, University of Turku, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | | | - Riitta Parkkola
- Department of Radiology, Turku University Hospital, University of Turku, Turku, Finland
| | - Mira Karrasch
- Department of Psychology, Åbo Akademi University, Turku, Finland
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Juha O Rinne
- Turku PET Centre, University of Turku, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| |
Collapse
|
42
|
Cousins KAQ, Irwin DJ, Chen-Plotkin A, Shaw LM, Arezoumandan S, Lee EB, Wolk DA, Weintraub D, Spindler M, Deik A, Grossman M, Tropea TF. Plasma GFAP associates with secondary Alzheimer's pathology in Lewy body disease. Ann Clin Transl Neurol 2023; 10:802-813. [PMID: 37000892 DOI: 10.1002/acn3.51768] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 04/03/2023] Open
Abstract
OBJECTIVE Within Lewy body spectrum disorders (LBSD) with α-synuclein pathology (αSyn), concomitant Alzheimer's disease (AD) pathology is common and is predictive of clinical outcomes, including cognitive impairment and decline. Plasma phosphorylated tau 181 (p-tau181 ) is sensitive to AD neuropathologic change (ADNC) in clinical AD, and plasma glial fibrillary acidic protein (GFAP) is associated with the presence of β-amyloid plaques. While these plasma biomarkers are well tested in clinical and pathological AD, their diagnostic and prognostic performance for concomitant AD in LBSD is unknown. METHODS In autopsy-confirmed αSyn-positive LBSD, we tested how plasma p-tau181 and GFAP differed across αSyn with concomitant ADNC (αSyn+AD; n = 19) and αSyn without AD (αSyn; n = 30). Severity of burden was scored on a semiquantitative scale for several pathologies (e.g., β-amyloid and tau), and scores were averaged across sampled brainstem, limbic, and neocortical regions. RESULTS Linear models showed that plasma GFAP was significantly higher in αSyn+AD compared to αSyn (β = 0.31, 95% CI = 0.065-0.56, and P = 0.015), after covarying for age at plasma, plasma-to-death interval, and sex; plasma p-tau181 was not (P = 0.37). Next, linear models tested associations of AD pathological features with both plasma analytes, covarying for plasma-to-death, age at plasma, and sex. GFAP was significantly associated with brain β-amyloid (β = 15, 95% CI = 6.1-25, and P = 0.0018) and tau burden (β = 12, 95% CI = 2.5-22, and P = 0.015); plasma p-tau181 was not associated with either (both P > 0.34). INTERPRETATION Findings indicate that plasma GFAP may be sensitive to concomitant AD pathology in LBSD, especially accumulation of β-amyloid plaques.
Collapse
Affiliation(s)
- Katheryn A Q Cousins
- Department of Neurology, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sanaz Arezoumandan
- Department of Neurology, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David A Wolk
- Department of Neurology, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Daniel Weintraub
- Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Meredith Spindler
- Department of Neurology, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Andres Deik
- Department of Neurology, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Murray Grossman
- Department of Neurology, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Thomas F Tropea
- Department of Neurology, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
43
|
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
|
44
|
Chun MY, Jang H, Kim HJ, Kim JP, Gallacher J, Allué JA, Sarasa L, Castillo S, Pascual-Lucas M, Na DL, Seo SW. Contribution of clinical information to the predictive performance of plasma β-amyloid levels for amyloid positron emission tomography positivity. Front Aging Neurosci 2023; 15:1126799. [PMID: 36998318 PMCID: PMC10044013 DOI: 10.3389/fnagi.2023.1126799] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/24/2023] [Indexed: 03/15/2023] Open
Abstract
BackgroundEarly detection of β-amyloid (Aβ) accumulation, a major biomarker for Alzheimer’s disease (AD), has become important. As fluid biomarkers, the accuracy of cerebrospinal fluid (CSF) Aβ for predicting Aβ deposition on positron emission tomography (PET) has been extensively studied, and the development of plasma Aβ is beginning to receive increased attention recently. In the present study, we aimed to determine whether APOE genotypes, age, and cognitive status increase the predictive performance of plasma Aβ and CSF Aβ levels for Aβ PET positivity.MethodsWe recruited 488 participants who underwent both plasma Aβ and Aβ PET studies (Cohort 1) and 217 participants who underwent both cerebrospinal fluid (CSF) Aβ and Aβ PET studies (Cohort 2). Plasma and CSF samples were analyzed using ABtest-MS, an antibody-free liquid chromatography-differential mobility spectrometry-triple quadrupole mass spectrometry method and INNOTEST enzyme-linked immunosorbent assay kits, respectively. To evaluate the predictive performance of plasma Aβ and CSF Aβ, respectively, logistic regression and receiver operating characteristic analyses were performed.ResultsWhen predicting Aβ PET status, both plasma Aβ42/40 ratio and CSF Aβ42 showed high accuracy (plasma Aβ area under the curve (AUC) 0.814; CSF Aβ AUC 0.848). In the plasma Aβ models, the AUC values were higher than plasma Aβ alone model, when the models were combined with either cognitive stage (p < 0.001) or APOE genotype (p = 0.011). On the other hand, there was no difference between the CSF Aβ models, when these variables were added.ConclusionPlasma Aβ might be a useful predictor of Aβ deposition on PET status as much as CSF Aβ, particularly when considered with clinical information such as APOE genotype and cognitive stage.
Collapse
Affiliation(s)
- Min Young Chun
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, Republic of Korea
| | - Hyemin Jang
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- *Correspondence: Hyemin Jang, ; Sang Won Seo,
| | - Hee Jin Kim
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
- Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Jun Pyo Kim
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Center for Neuroimaging, Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
| | - John Gallacher
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
| | | | | | | | | | - Duk L. Na
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Sang Won Seo
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
- Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
- Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
- *Correspondence: Hyemin Jang, ; Sang Won Seo,
| | | |
Collapse
|
45
|
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
|
46
|
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
|
47
|
Wan K, Yin W, Tang Y, Zhu W, Wang Z, Zhou X, Zhang W, Zhang C, Yu X, Zhao W, Li C, Zhu X, Sun Z. Brain Gray Matter Volume Mediated the Correlation Between Plasma P-Tau and Cognitive Function of Early Alzheimer's Disease in China: A Cross-Sectional Observational Study. J Alzheimers Dis 2023; 92:81-93. [PMID: 36710682 DOI: 10.3233/jad-221100] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND The primary manifestations of Alzheimer's disease (AD) include cognitive decline and brain gray matter volume (GMV) atrophy. Recent studies have found that plasma phosphorylated-tau (p-tau) concentrations perform better in diagnosing, differentiating, and monitoring the progression of AD. However, the correlation between plasma p-tau, GMV, and cognition remains unclear. OBJECTIVE To investigate whether GMV plays a mediating role in the association between plasma p-tau concentrations and cognition. METHODS In total, 99 participants (47 patients with AD and 52 cognitively unimpaired [CU] individuals) were included. All participants underwent neuropsychological assessments, laboratory examinations, and magnetic resonance imaging scans. Plasma p-tau217 and p-tau181 concentrations were measured using an enzyme-linked immunosorbent assay kit. Voxel-based morphometry was performed to assess participants' brain GMV. Partial correlation and mediation analyses were conducted in AD group. RESULTS Plasma p-tau concentrations were significantly higher in the AD group than in the CU group. Patients with AD had significant brain GMV atrophy in the right hippocampus, bilateral middle temporal gyrus, and right inferior temporal gyrus. In the AD group, there were significant correlations between plasma p-tau217 concentrations, GMV, and Mini-Mental State Examination (MMSE) scores. Brain GMV of the right hippocampus mediated the association between plasma p-tau217 concentrations and MMSE scores. A significant correlation between plasma p-tau181 and MMSE scores was not identified. CONCLUSION The findings indicate that p-tau217 is a promising biomarker for central processes affecting brain GMV and cognitive function. This may provide potential targets for future intervention and treatment of tau-targeting therapies in the early stages of AD.
Collapse
Affiliation(s)
- Ke Wan
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Wenwen Yin
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Yating Tang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Wenhao Zhu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Zhiqiang Wang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Menzies Institute for Medical Research, University of Tasmania, Private Bag 23, Hobart, Tasmania, Australia
| | - Xia Zhou
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Wei Zhang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Cun Zhang
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xianfeng Yu
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Wenming Zhao
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chenchen Li
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Xiaoqun Zhu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Zhongwu Sun
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| |
Collapse
|
48
|
Tropea TF, Waligorska T, Xie SX, Nasrallah IM, Cousins KAQ, Trojanowski JQ, Grossman M, Irwin DJ, Weintraub D, Lee EB, Wolk DA, Chen-Plotkin AS, Shaw LM. Plasma phosphorylated tau181 predicts cognitive and functional decline. Ann Clin Transl Neurol 2023; 10:18-31. [PMID: 36518085 PMCID: PMC9852389 DOI: 10.1002/acn3.51695] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 10/11/2022] [Accepted: 10/31/2022] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE To determine if plasma tau phosphorylated at threonine 181 (p-tau181) distinguishes pathology-confirmed Alzheimer's disease (AD) from normal cognition (NC) adults, to test if p-tau181 predicts cognitive and functional decline, and to validate findings in an external cohort. METHODS Thirty-one neuropathology-confirmed AD cases, participants with clinical diagnoses of mild cognitive impairment (MCI, N = 91) or AD dementia (N = 64), and NC (N = 241) had plasma collected at study entry. The clinical diagnosis groups had annual cognitive (Mini-Mental State Examination, MMSE) and functional (Clinical Dementia Rating Scale, CDR) measures. NC (N = 70), MCI (N = 75), and AD dementia (N = 50) cases from the Alzheimer's Disease Neuroimaging Initiative (ADNI) were used as a validation cohort. Plasma p-tau181 was measured using the Quanterix SiMoA HD-X platform. RESULTS Plasma p-tau181 differentiated pathology-confirmed AD from NC with negative amyloid PET scans with an AUC of 0.93. A cut point of 3.44 pg/mL (maximum Youden Index) had a sensitivity of 0.77, specificity of 0.96. p-Tau181 values above the cut point were associated with the faster rate of decline in MMSE in AD dementia and MCI and a shorter time to a clinically significant functional decline in all groups. In a subset of MCI cases from ADNI, p-tau181 values above the cut point associated with faster rate of decline in MMSE, and a shorter time to a clinically significant functional decline and conversion to dementia. INTERPRETATION Plasma p-tau181 differentiates AD pathology cases from NC with high accuracy. Higher levels of plasma p-tau181 are associated with faster cognitive and functional decline.
Collapse
Affiliation(s)
- Thomas F Tropea
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Teresa Waligorska
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sharon X Xie
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ilya M Nasrallah
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Katheryn A Q Cousins
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Murray Grossman
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel Weintraub
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Parkinson's Disease Research, Education and Clinical Center (PADRECC), Philadelphia Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David A Wolk
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alice S Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
49
|
Ashton NJ, Janelidze S, Mattsson-Carlgren N, Binette AP, Strandberg O, Brum WS, Karikari TK, González-Ortiz F, Di Molfetta G, Meda FJ, Jonaitis EM, Koscik RL, Cody K, Betthauser TJ, Li Y, Vanmechelen E, Palmqvist S, Stomrud E, Bateman RJ, Zetterberg H, Johnson SC, Blennow K, Hansson O. Differential roles of Aβ42/40, p-tau231 and p-tau217 for Alzheimer's trial selection and disease monitoring. Nat Med 2022; 28:2555-2562. [PMID: 36456833 PMCID: PMC9800279 DOI: 10.1038/s41591-022-02074-w] [Citation(s) in RCA: 195] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 10/03/2022] [Indexed: 12/03/2022]
Abstract
Blood biomarkers indicative of Alzheimer's disease (AD) pathology are altered in both preclinical and symptomatic stages of the disease. Distinctive biomarkers may be optimal for the identification of AD pathology or monitoring of disease progression. Blood biomarkers that correlate with changes in cognition and atrophy during the course of the disease could be used in clinical trials to identify successful interventions and thereby accelerate the development of efficient therapies. When disease-modifying treatments become approved for use, efficient blood-based biomarkers might also inform on treatment implementation and management in clinical practice. In the BioFINDER-1 cohort, plasma phosphorylated (p)-tau231 and amyloid-β42/40 ratio were more changed at lower thresholds of amyloid pathology. Longitudinally, however, only p-tau217 demonstrated marked amyloid-dependent changes over 4-6 years in both preclinical and symptomatic stages of the disease, with no such changes observed in p-tau231, p-tau181, amyloid-β42/40, glial acidic fibrillary protein or neurofilament light. Only longitudinal increases of p-tau217 were also associated with clinical deterioration and brain atrophy in preclinical AD. The selective longitudinal increase of p-tau217 and its associations with cognitive decline and atrophy was confirmed in an independent cohort (Wisconsin Registry for Alzheimer's Prevention). These findings support the differential association of plasma biomarkers with disease development and strongly highlight p-tau217 as a surrogate marker of disease progression in preclinical and prodromal AD, with impact for the development of new disease-modifying treatments.
Collapse
Affiliation(s)
- Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Shorena Janelidze
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
| | - Wagner S Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Fernándo González-Ortiz
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Guglielmo Di Molfetta
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Francisco J Meda
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Erin M Jonaitis
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Rebecca Langhough Koscik
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Karly Cody
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Tobey J Betthauser
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Sebastian Palmqvist
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- 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
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
- ADx NeuroSciences, Technologiepark 94, Ghent, Belgium.
| | - Oskar Hansson
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
50
|
Wilson EN, Young CB, Ramos Benitez J, Swarovski MS, Feinstein I, Vandijck M, Le Guen Y, Kasireddy NM, Shahid M, Corso NK, Wang Q, Kennedy G, Trelle AN, Lind B, Channappa D, Belnap M, Ramirez V, Skylar-Scott I, Younes K, Yutsis MV, Le Bastard N, Quinn JF, van Dyck CH, Nairn A, Fredericks CA, Tian L, Kerchner GA, Montine TJ, Sha SJ, Davidzon G, Henderson VW, Longo FM, Greicius MD, Wagner AD, Wyss-Coray T, Poston KL, Mormino EC, Andreasson KI. Performance of a fully-automated Lumipulse plasma phospho-tau181 assay for Alzheimer's disease. Alzheimers Res Ther 2022; 14:172. [PMID: 36371232 PMCID: PMC9652927 DOI: 10.1186/s13195-022-01116-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/31/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND The recent promise of disease-modifying therapies for Alzheimer's disease (AD) has reinforced the need for accurate biomarkers for early disease detection, diagnosis and treatment monitoring. Advances in the development of novel blood-based biomarkers for AD have revealed that plasma levels of tau phosphorylated at various residues are specific and sensitive to AD dementia. However, the currently available tests have shortcomings in access, throughput, and scalability that limit widespread implementation. METHODS We evaluated the diagnostic and prognostic performance of a high-throughput and fully-automated Lumipulse plasma p-tau181 assay for the detection of AD. Plasma from older clinically unimpaired individuals (CU, n = 463) and patients with mild cognitive impairment (MCI, n = 107) or AD dementia (n = 78) were obtained from the longitudinal Stanford University Alzheimer's Disease Research Center (ADRC) and the Stanford Aging and Memory Study (SAMS) cohorts. We evaluated the discriminative accuracy of plasma p-tau181 for clinical AD diagnosis, association with amyloid β peptides and p-tau181 concentrations in CSF, association with amyloid positron emission tomography (PET), and ability to predict longitudinal cognitive and functional change. RESULTS The assay showed robust performance in differentiating AD from control participants (AUC 0.959, CI: 0.912 to 0.990), and was strongly associated with CSF p-tau181, CSF Aβ42/Aβ40 ratio, and amyloid-PET global SUVRs. Associations between plasma p-tau181 with CSF biomarkers were significant when examined separately in Aβ+ and Aβ- groups. Plasma p-tau181 significantly increased over time in CU and AD diagnostic groups. After controlling for clinical diagnosis, age, sex, and education, baseline plasma p-tau181 predicted change in MoCA overall and change in CDR Sum of Boxes in the AD group over follow-up of up to 5 years. CONCLUSIONS This fully-automated and available blood-based biomarker assay therefore may be useful for early detection, diagnosis, prognosis, and treatment monitoring of AD.
Collapse
Affiliation(s)
- Edward N. Wilson
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA
| | - Christina B. Young
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Javier Ramos Benitez
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Michelle S. Swarovski
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Igor Feinstein
- grid.168010.e0000000419368956Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA USA
| | | | - Yann Le Guen
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Nandita M. Kasireddy
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Marian Shahid
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Nicole K. Corso
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Qian Wang
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Gabriel Kennedy
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Alexandra N. Trelle
- grid.168010.e0000000419368956Psychology, Stanford University, Stanford, CA USA
| | - Betty Lind
- grid.410404.50000 0001 0165 2383Neurology, Portland VA Medical Center, Portland, OR USA ,grid.5288.70000 0000 9758 5690Neurology, Oregon Health & Science University, Portland, OR USA
| | - Divya Channappa
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Pathology, Stanford University, Stanford, CA USA
| | - Malia Belnap
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Veronica Ramirez
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Irina Skylar-Scott
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Kyan Younes
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Maya V. Yutsis
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | | | - Joseph F. Quinn
- grid.410404.50000 0001 0165 2383Neurology, Portland VA Medical Center, Portland, OR USA ,grid.5288.70000 0000 9758 5690Neurology, Oregon Health & Science University, Portland, OR USA
| | | | - Angus Nairn
- grid.47100.320000000419368710Psychiatry, Yale University, New Haven, CT USA
| | - Carolyn A. Fredericks
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Lu Tian
- grid.168010.e0000000419368956Biomedical Data Science, Stanford University, Stanford, CA USA
| | - Geoffrey A. Kerchner
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Thomas J. Montine
- grid.168010.e0000000419368956Pathology, Stanford University, Stanford, CA USA
| | - Sharon J. Sha
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Guido Davidzon
- grid.168010.e0000000419368956Radiology, Stanford University, Stanford, CA USA
| | - Victor W. Henderson
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Epidemiology & Population Health, Stanford University, Stanford, CA USA
| | - Frank M. Longo
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA
| | - Michael D. Greicius
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA
| | - Anthony D. Wagner
- grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Psychology, Stanford University, Stanford, CA USA
| | - Tony Wyss-Coray
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA
| | - Kathleen L. Poston
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA
| | - Elizabeth C. Mormino
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA
| | - Katrin I. Andreasson
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA ,grid.499295.a0000 0004 9234 0175Chan Zuckerberg Biohub, San Francisco, CA 94158 USA
| |
Collapse
|