51
|
Heshmatollah A, Fani L, Koudstaal PJ, Ghanbari M, Ikram MA, Ikram MK. Plasma Amyloid Beta, Total-Tau and Neurofilament Light Chain Levels and the Risk of Stroke: A Prospective Population-Based Study. Neurology 2022; 98:e1729-e1737. [PMID: 35232820 DOI: 10.1212/wnl.0000000000200004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 01/03/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES To unravel whether Alzheimer's disease-related pathology or neurodegeneration play a role in stroke etiology, we determined the effect of plasma levels amyloid β (Aβ), total-tau and neurofilament light chain (NfL) on risk of stroke and its subtypes. METHODS Between 2002 and 2005, we measured plasma Aβ40, Aβ42, total-tau, and NfL in 4,661 stroke-free participants from the population-based Rotterdam Study. We used Cox proportional-hazards models to determine the association between these markers with incident stroke for the entire cohort, per stroke subtype, and by median age, sex, Apolipoprotein E (APOE) ε4 carriership, and education. RESULTS After a mean follow-up of 10.8 ± 3.3 years, 379 participants suffered a first-ever stroke. Log2 total-tau at baseline showed a non-linear association with risk of any stroke and ischemic stroke: compared to the first (lowest) quartile the adjusted hazard ratio for the highest quartile total-tau was 1.68, 95% CI: 1.18-2.40 for any stroke. Log2 NfL was associated with an increased risk of any stroke (HR per SD increase 1.27, 95% CI: 1.12-1.44), ischemic stroke, and hemorrhagic stroke (HR 1.56, 95% CI: 1.14-2.12). Log2 Aβ40, Aβ42, and Aβ42/40 ratio levels were not associated with stroke risk.Discussion Participants with higher total-tau and NfL at baseline had a higher risk of stroke and several stroke subtypes. These findings support the role of markers of neurodegeneration in the etiology of stroke. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that higher plasma levels of total-tau and NfL are associated with an increased risk of subsequent stroke.
Collapse
Affiliation(s)
- Alis Heshmatollah
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands.,Department of Neurology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Lana Fani
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Peter J Koudstaal
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - M Kamran Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands.,Department of Neurology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
52
|
Álvarez-Sánchez L, Peña-Bautista C, Baquero M, Cháfer-Pericás C. Novel Ultrasensitive Detection Technologies for the Identification of Early and Minimally Invasive Alzheimer's Disease Blood Biomarkers. J Alzheimers Dis 2022; 86:1337-1369. [PMID: 35213367 DOI: 10.3233/jad-215093] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Single molecule array (SIMOA) and other ultrasensitive detection technologies have allowed the determination of blood-based biomarkers of Alzheimer's disease (AD) for diagnosis and monitoring, thereby opening up a promising field of research. OBJECTIVE To review the published bibliography on plasma biomarkers in AD using new ultrasensitive techniques. METHODS A systematic review of the PubMed database was carried out to identify reports on the use of blood-based ultrasensitive technology to identify biomarkers for AD. RESULTS Based on this search, 86 works were included and classified according to the biomarker determined. First, plasma amyloid-β showed satisfactory accuracy as an AD biomarker in patients with a high risk of developing dementia. Second, plasma t-Tau displayed good sensitivity in detecting different neurodegenerative diseases. Third, plasma p-Tau was highly specific for AD. Fourth, plasma NfL was highly sensitive for distinguishing between patients with neurodegenerative diseases and healthy controls. In general, the simultaneous determination of several biomarkers facilitated greater accuracy in diagnosing AD (Aβ42/Aβ40, p-Tau181/217). CONCLUSION The recent development of ultrasensitive technology allows the determination of blood-based biomarkers with high sensitivity, thus facilitating the early detection of AD through the analysis of easily obtained biological samples. In short, as a result of this knowledge, pre-symptomatic and early AD diagnosis may be possible, and the recruitment process for future clinical trials could be more precise. However, further studies are necessary to standardize levels of blood-based biomarkers in the general population and thus achieve reproducible results among different laboratories.
Collapse
Affiliation(s)
| | - Carmen Peña-Bautista
- Alzheimer Disease Research Group, Health Research Institute La Fe, Valencia, Spain
| | - Miguel Baquero
- Division of Neurology, University and Polytechnic Hospital La Fe, Valencia, Spain
| | | |
Collapse
|
53
|
Tau proteins in blood as biomarkers of Alzheimer's disease and other proteinopathies. J Neural Transm (Vienna) 2022; 129:239-259. [PMID: 35175385 DOI: 10.1007/s00702-022-02471-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/02/2022] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD), the most common age-dependent neurodegenerative disorder, is characterized neuropathologically by extracellular Aβ plaques and intracellular tau neurofibrillary tangles. While in AD tau pathology probably follows early alterations in Aβ metabolism, it develops independently in the so-called primary tauopathies, the main form being frontotemporal lobar degeneration with tau pathology. Tau pathology in AD brain is reflected in the cerebrospinal fluid (CSF) by elevated levels of the two AD tau biomarkers total and phosphorylated tau, which are now used for routine diagnostic purposes. On the contrary, no established neurochemical biomarkers exist for tau pathology in primary tauopathies. Thanks to recent technological advances, total and phosphorylated tau can now be quantified also on peripheral blood, and accumulating evidence shows that measurement of plasma phosphorylated tau species (P-tau181, P-tau217, and P-tau231) has high performances in discriminating AD patients from cognitively unimpaired subjects but also from patients with other dementias. Moreover, plasma P-tau levels are associated with tracer uptake on tau- and amyloid-PET as well as with brain atrophy, cognitive measures and longitudinal changes of these parameters. These features, together with the low invasiveness, scalability, and ease of longitudinal sampling, which differentiate plasma P-tau species from their CSF counterparts, make these proteins promising peripheral biomarkers for AD in both research and clinical setting. This review discusses the recent developments in the field of plasma tau proteins as diagnostic, pathophysiological and prognostic biomarkers of Alzheimer's disease; additional findings from the fields of genetic forms of AD and of non-AD proteinopathies are also summarized.
Collapse
|
54
|
Tissot C, Therriault J, Kunach P, L Benedet A, Pascoal TA, Ashton NJ, Karikari TK, Servaes S, Lussier FZ, Chamoun M, Tudorascu DL, Stevenson J, Rahmouni N, Poltronetti NM, Pallen V, Bezgin G, Kang MS, Mathotaarachchi SS, Wang YT, Fernandez Arias J, Ferreira PCL, Ferrari-Souza JP, Vanmechelen E, Blennow K, Zetterberg H, Gauthier S, Rosa-Neto P. Comparing tau status determined via plasma pTau181, pTau231 and [ 18F]MK6240 tau-PET. EBioMedicine 2022; 76:103837. [PMID: 35134647 PMCID: PMC8844756 DOI: 10.1016/j.ebiom.2022.103837] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/24/2021] [Accepted: 01/11/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Tau in Alzheimer's disease (AD) is assessed via cerebrospinal fluid (CSF) and Positron emission tomography (PET). Novel methods to detect phosphorylated tau (pTau) in blood have been recently developed. We aim to investigate agreement of tau status as determined by [18F]MK6240 tau-PET, plasma pTau181 and pTau231. METHODS We assessed cognitively unimpaired young, cognitively unimpaired, mild cognitive impairment and AD individuals with [18F]MK6240, plasma pTau181, pTau 231, [18F]AZD4694 amyloid-PET and MRI. A subset underwent CSF assessment. We conducted ROC curves to obtain cut-off values for plasma pTau epitopes. Individuals were categorized as positive or negative in all biomarkers. We then compared the distribution among concordant and discordant groups in relation to diagnosis, Aβ status, APOEε4 status, [18F]AZD4694 global SUVR, hippocampal volume and CSF pTau181. FINDINGS The threshold for positivity was 15.085 pg/mL for plasma pTau181 and 17.652 pg/mL for plasma pTau231. Most individuals had concordant statuses, however, 18% of plasma181/PET, 26% of plasma231/PET and 25% of the pTau231/pTau181 were discordant. Positivity to at least one biomarker was often accompanied by diagnosis of cognitive impairment, Aβ positivity, APOEε4 carriership, higher levels of [18F]AZD4694 global SUVR, hippocampal atrophy and CSF pTau181. INTERPRETATION Plasma pTau181, pTau231 and [18F]MK6240 seem to reflect different stages of tau progression. Plasma biomarkers can be useful in the context of diagnostic information and clinical trials, to evaluate the disease stage. Moreover, they seem to confidently evaluate tau-PET positivity. FUNDING Moreover, this study was supported by Weston Brain Institute, Canadian Institute of Health Research and Fonds de Recherche du Québec.
Collapse
Affiliation(s)
- Cécile Tissot
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Joseph Therriault
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Peter Kunach
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Andréa L Benedet
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Tharick A Pascoal
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada; University of Pittsburgh, Pittsburgh, PA, USA
| | - Nicholas J Ashton
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Sweden; King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl 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
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Sweden; University of Pittsburgh, Pittsburgh, PA, USA
| | - Stijn Servaes
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Firoza Z Lussier
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Mira Chamoun
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | | | - Jenna Stevenson
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Nesrine Rahmouni
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Nina Margherita Poltronetti
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Vanessa Pallen
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Gleb Bezgin
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Min Su Kang
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Sulantha S Mathotaarachchi
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Yi-Ting Wang
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Jaime Fernandez Arias
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | | | - João Pedro Ferrari-Souza
- University of Pittsburgh, Pittsburgh, PA, USA; Graduate program in Biological Sciences, Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; UK Dementia Research Institute at UCL, London, United Kingdom
| | - Henrik Zetterberg
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Sweden; UK Dementia Research Institute at UCL, London, United Kingdom; Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Serge Gauthier
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Douglas Hospital Research Centre, Verdun, QC, Canada
| | - Pedro Rosa-Neto
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada.
| |
Collapse
|
55
|
Kim Y, Kim J, Son M, Lee J, Yeo I, Choi KY, Kim H, Kim BC, Lee KH, Kim Y. Plasma protein biomarker model for screening Alzheimer disease using multiple reaction monitoring-mass spectrometry. Sci Rep 2022; 12:1282. [PMID: 35075217 PMCID: PMC8786819 DOI: 10.1038/s41598-022-05384-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 01/11/2022] [Indexed: 12/01/2022] Open
Abstract
Alzheimer disease (AD) is a leading cause of dementia that has gained prominence in our aging society. Yet, the complexity of diagnosing AD and measuring its invasiveness poses an obstacle. To this end, blood-based biomarkers could mitigate the inconveniences that impede an accurate diagnosis. We developed models to diagnose AD and measure the severity of neurocognitive impairment using blood protein biomarkers. Multiple reaction monitoring-mass spectrometry, a highly selective and sensitive approach for quantifying targeted proteins in samples, was used to analyze blood samples from 4 AD groups: cognitive normal control, asymptomatic AD, prodromal AD), and AD dementia. Multimarker models were developed using 10 protein biomarkers and apolipoprotein E genotypes for amyloid beta and 10 biomarkers with Korean Mini-Mental Status Examination (K-MMSE) score for predicting Alzheimer disease progression. The accuracies for the AD classification model and AD progression monitoring model were 84.9% (95% CI 82.8 to 87.0) and 79.1% (95% CI 77.8 to 80.5), respectively. The models were more accurate in diagnosing AD, compared with single APOE genotypes and the K-MMSE score. Our study demonstrates the possibility of predicting AD with high accuracy by blood biomarker analysis as an alternative method of screening for AD.
Collapse
Affiliation(s)
- Yeongshin Kim
- Interdisciplinary Program of Bioengineering, Seoul National University College of Engineering, Seoul, Republic of Korea
| | - Jaenyeon Kim
- Interdisciplinary Program of Bioengineering, Seoul National University College of Engineering, Seoul, Republic of Korea
| | - Minsoo Son
- Interdisciplinary Program of Bioengineering, Seoul National University College of Engineering, Seoul, Republic of Korea
| | - Jihyeon Lee
- Department of Biomedical Engineering, Seoul National University College of Medicine, 28 Yongon-Dong, Chongno-Ku, Seoul, 110-799, Republic of Korea
| | - Injoon Yeo
- Department of Biomedical Engineering, Seoul National University College of Medicine, 28 Yongon-Dong, Chongno-Ku, Seoul, 110-799, Republic of Korea
| | - Kyu Yeong Choi
- Gwangju Alzheimer's Disease and Related Dementia Cohort Research Center and Department of Biomedical Science, Chosun University, Gwangju, 61452, Republic of Korea
| | - Hoowon Kim
- Gwangju Alzheimer's Disease and Related Dementia Cohort Research Center and Department of Biomedical Science, Chosun University, Gwangju, 61452, Republic of Korea
- Department of Neurology, Chosun University Hospital, Gwangju, 61452, Republic of Korea
| | - Byeong C Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Kun Ho Lee
- Gwangju Alzheimer's Disease and Related Dementia Cohort Research Center and Department of Biomedical Science, Chosun University, Gwangju, 61452, Republic of Korea.
- Department of Biomedical Science, Chosun University, Gwangju, 61452, Republic of Korea.
- Aging Neuroscience Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea.
| | - Youngsoo Kim
- Interdisciplinary Program of Bioengineering, Seoul National University College of Engineering, Seoul, Republic of Korea.
- Department of Biomedical Engineering, Seoul National University College of Medicine, 28 Yongon-Dong, Chongno-Ku, Seoul, 110-799, Republic of Korea.
| |
Collapse
|
56
|
Eldem E, Barve A, Sallin O, Foucras S, Annoni JM, Schmid AW, Alberi Auber L. Salivary Proteomics Identifies Transthyretin as a Biomarker of Early Dementia Conversion. J Alzheimers Dis Rep 2022; 6:31-41. [PMID: 35360272 PMCID: PMC8925122 DOI: 10.3233/adr-210056] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/06/2022] [Indexed: 01/18/2023] Open
Abstract
Background: Alzheimer’s disease (AD) remains to date an incurable disease with a long asymptomatic phase. Early diagnosis in peripheral biofluids has emerged as key for identifying subjects at risk and developing therapeutics and preventative approaches. Objective: We apply proteomics discovery to identify salivary diagnostic biomarkers for AD, which are suitable for self-sampling and longitudinal biomonitoring during aging. Methods: 57 participants were recruited for the study and were categorized into Cognitively normal (CNh) (n = 19), mild cognitive impaired (MCI) (n = 21), and Alzheimer’s disease (AD) (n = 17). On a subset of subjects, 3 CNh and 3 mild AD, shot-gun filter aided sample preparation (FASP) proteomics and liquid chromatography mass spectroscopy (LC-MS/MS) was employed in saliva and cerebrospinal fluid (CSF) to identify neural-derived proteins. The protein level of salivary Transthyretin (TTR) was validated using western blot analysis across groups. Results: We found that 19.8% of the proteins in saliva are shared with CSF. When we compared the saliva and CSF proteome, 24 hits were decreased with only one protein expressed more. Among the differentially expressed proteins, TTR with reported function in amyloid misfolding, shows a significant drop in AD samples, confirmed by western blot showing a 0.5-fold reduction in MCI and AD compared to CNh. Conclusion: A reduction in salivary TTR appears with the onset of cognitive symptoms. More in general, the proteomic profiling of saliva shows a plethora of biomarkers worth pursuing as non-invasive hallmarks of dementia in the preclinical stage.
Collapse
Affiliation(s)
- Ece Eldem
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland
- Swiss Integrative Center for Human Health, Fribourg, Switzerland
| | - Aatmika Barve
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland
- Swiss Integrative Center for Human Health, Fribourg, Switzerland
| | - Olivier Sallin
- Swiss Integrative Center for Human Health, Fribourg, Switzerland
| | | | - Jean-Marie Annoni
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland
- Hôpital Cantonal Fribourgeois, Fribourg, Switzerland
| | | | - Lavinia Alberi Auber
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland
- Swiss Integrative Center for Human Health, Fribourg, Switzerland
| |
Collapse
|
57
|
Leuzy A, Mattsson‐Carlgren N, Palmqvist S, Janelidze S, Dage JL, Hansson O. Blood-based biomarkers for Alzheimer's disease. EMBO Mol Med 2022; 14:e14408. [PMID: 34859598 PMCID: PMC8749476 DOI: 10.15252/emmm.202114408] [Citation(s) in RCA: 137] [Impact Index Per Article: 68.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 11/02/2021] [Accepted: 11/05/2021] [Indexed: 12/01/2022] Open
Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD) represent a mounting public health challenge. As these diseases are difficult to diagnose clinically, biomarkers of underlying pathophysiology are playing an ever-increasing role in research, clinical trials, and in the clinical work-up of patients. Though cerebrospinal fluid (CSF) and positron emission tomography (PET)-based measures are available, their use is not widespread due to limitations, including high costs and perceived invasiveness. As a result of rapid advances in the development of ultra-sensitive assays, the levels of pathological brain- and AD-related proteins can now be measured in blood, with recent work showing promising results. Plasma P-tau appears to be the best candidate marker during symptomatic AD (i.e., prodromal AD and AD dementia) and preclinical AD when combined with Aβ42/Aβ40. Though not AD-specific, blood NfL appears promising for the detection of neurodegeneration and could potentially be used to detect the effects of disease-modifying therapies. This review provides an overview of the progress achieved thus far using AD blood-based biomarkers, highlighting key areas of application and unmet challenges.
Collapse
Affiliation(s)
- Antoine Leuzy
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityMalmöSweden
| | - Niklas Mattsson‐Carlgren
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityMalmöSweden
- Department of NeurologySkåne University HospitalLundSweden
- Wallenberg Centre for Molecular MedicineLund UniversityLundSweden
| | - Sebastian Palmqvist
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityMalmöSweden
- Memory ClinicSkåne University HospitalLundSweden
| | - Shorena Janelidze
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityMalmöSweden
| | - Jeffrey L Dage
- Stark Neuroscience Research InstituteIndiana University School of MedicineIndianapolisINUSA
| | - Oskar Hansson
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityMalmöSweden
- Memory ClinicSkåne University HospitalLundSweden
| |
Collapse
|
58
|
Achard V, Ceyzériat K, Tournier BB, Frisoni GB, Garibotto V, Zilli T. Biomarkers to Evaluate Androgen Deprivation Therapy for Prostate Cancer and Risk of Alzheimer's Disease and Neurodegeneration: Old Drugs, New Concerns. Front Oncol 2022; 11:734881. [PMID: 34970480 PMCID: PMC8712866 DOI: 10.3389/fonc.2021.734881] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/15/2021] [Indexed: 11/29/2022] Open
Abstract
Androgen deprivation therapy (ADT) is a standard treatment for prostate cancer patients, routinely used in the palliative or in the curative setting in association with radiotherapy. Among the systemic long-term side effects of ADT, growing data suggest a potentially increased risk of dementia/Alzheimer’s disease in prostate cancer patients treated with hormonal manipulation. While pre-clinical data suggest that androgen ablation may have neurotoxic effects due to Aβ accumulation and increased tau phosphorylation in small animal brains, clinical studies have measured the impact of ADT on long-term cognitive function, with conflicting results, and studies on biological changes after ADT are still lacking. The aim of this review is to report on the current evidence on the association between the ADT use and the risk of cognitive impairment in prostate cancer patients. We will focus on the contribution of Alzheimer’s disease biomarkers, namely through imaging, to investigate potential ADT-induced brain modifications. The evidence from these preliminary studies shows brain changes in gray matter volume, cortical activation and metabolism associated with ADT, however with a large variability in biomarker selection, ADT duration and cognitive outcome. Importantly, no study investigated yet biomarkers of Alzheimer’s disease pathology, namely amyloid and tau. These preliminary data emphasize the need for larger targeted investigations.
Collapse
Affiliation(s)
- Vérane Achard
- Division of Radiation Oncology, Department of Oncology, Geneva University Hospitals and Faculty of Medicine, Geneva University, Geneva, Switzerland
| | - Kelly Ceyzériat
- Division of Radiation Oncology, Department of Oncology, Geneva University Hospitals and Faculty of Medicine, Geneva University, Geneva, Switzerland.,Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, Geneva University Hospitals, and NimtLab, Faculty of Medicine, Geneva University, Geneva, Switzerland.,Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals and Faculty of Medicine, Geneva University, Geneva, Switzerland
| | - Benjamin B Tournier
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals and Faculty of Medicine, Geneva University, Geneva, Switzerland
| | - Giovanni B Frisoni
- Memory Clinic, Department of Rehabilitation and Geriatrics, Geneva University and University Hospitals, Geneva, Switzerland
| | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, Geneva University Hospitals, and NimtLab, Faculty of Medicine, Geneva University, Geneva, Switzerland
| | - Thomas Zilli
- Division of Radiation Oncology, Department of Oncology, Geneva University Hospitals and Faculty of Medicine, Geneva University, Geneva, Switzerland
| |
Collapse
|
59
|
Abstract
With the expected rise in Alzheimer's disease and related dementias (ADRD) in the coming decades due to the aging population and a lack of effective disease-modifying treatments, there is a need for preventive strategies that may tap into resilience parameters. A wide array of resilience strategies has been proposed including genetics, socioeconomic status, lifestyle modifications, behavioral changes, and management of comorbid disease. These different strategies can be broadly classified as distinguishing between modifiable and non-modifiable risk factors, some of which can be quantified so that their clinical intervention can be effectively accomplished. A clear shift in research focus from dementia risk to addressing disease resistance and resilience is emerging that has provided new potential therapeutic targets. Here we review and summarize the latest investigations of resilience mechanisms and methods of quantifying resilience for clinical research. These approaches include identifying genetic variants that may help identify novel pathways (e.g., lipid metabolism, cellular trafficking, synaptic function, inflammation) for therapeutic treatments and biomarkers for use in a precision medicine-like regimen. In addition, innovative structural and molecular neuroimaging analyses may assist in detecting and quantifying pathological changes well before the onset of clinical symptoms setting up the possibility of primary and secondary prevention trials. Lastly, we summarize recent studies demonstrating the study of resilience in caregivers of persons living with dementia may have direct and indirect impact on the quality of care and patient outcomes.
Collapse
Affiliation(s)
- Mahesh S. Joshi
- Comprehensive Center for Brain Health, Department of Neurology, University of Miami Miller School of Medicine, Boca Raton, FL, USA
| | - James E. Galvin
- Comprehensive Center for Brain Health, Department of Neurology, University of Miami Miller School of Medicine, Boca Raton, FL, USA
| |
Collapse
|
60
|
Pathak N, Vimal SK, Tandon I, Agrawal L, Hongyi C, Bhattacharyya S. Neurodegenerative Disorders of Alzheimer, Parkinsonism, Amyotrophic Lateral Sclerosis and Multiple Sclerosis: An Early Diagnostic Approach for Precision Treatment. Metab Brain Dis 2022; 37:67-104. [PMID: 34719771 DOI: 10.1007/s11011-021-00800-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/11/2021] [Indexed: 12/21/2022]
Abstract
Neurodegenerative diseases (NDs) are characterised by progressive dysfunction of synapses, neurons, glial cells and their networks. Neurodegenerative diseases can be classified according to primary clinical features (e.g., dementia, parkinsonism, or motor neuron disease), anatomic distribution of neurodegeneration (e.g., frontotemporal degenerations, extrapyramidal disorders, or spinocerebellar degenerations), or principal molecular abnormalities. The most common neurodegenerative disorders are amyloidosis, tauopathies, a-synucleinopathy, and TAR DNA-binding protein 43 (TDP-43) proteopathy. The protein abnormalities in these disorders have abnormal conformational properties along with altered cellular mechanisms, and they exhibit motor deficit, mitochondrial malfunction, dysfunctions in autophagic-lysosomal pathways, synaptic toxicity, and more emerging mechanisms such as the roles of stress granule pathways and liquid-phase transitions. Finally, for each ND, microglial cells have been reported to be implicated in neurodegeneration, in particular, because the microglial responses can shift from neuroprotective to a deleterious role. Growing experimental evidence suggests that abnormal protein conformers act as seed material for oligomerization, spreading from cell to cell through anatomically connected neuronal pathways, which may in part explain the specific anatomical patterns observed in brain autopsy sample. In this review, we mention the human pathology of select neurodegenerative disorders, focusing on how neurodegenerative disorders (i.e., Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis) represent a great healthcare problem worldwide and are becoming prevalent because of the increasing aged population. Despite many studies have focused on their etiopathology, the exact cause of these diseases is still largely unknown and until now with the only available option of symptomatic treatments. In this review, we aim to report the systematic and clinically correlated potential biomarker candidates. Although future studies are necessary for their use in early detection and progression in humans affected by NDs, the promising results obtained by several groups leads us to this idea that biomarkers could be used to design a potential therapeutic approach and preclinical clinical trials for the treatments of NDs.
Collapse
Affiliation(s)
- Nishit Pathak
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China
| | - Sunil Kumar Vimal
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China
| | - Ishi Tandon
- Amity University Jaipur, Rajasthan, Jaipur, Rajasthan, India
| | - Lokesh Agrawal
- Graduate School of Comprehensive Human Sciences, Kansei Behavioural and Brain Sciences, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Cao Hongyi
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China
| | - Sanjib Bhattacharyya
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China.
| |
Collapse
|
61
|
Tahtouh T, Durieu E, Villiers B, Bruyère C, Nguyen TL, Fant X, Ahn KH, Khurana L, Deau E, Lindberg MF, Sévère E, Miege F, Roche D, Limanton E, L'Helgoual'ch JM, Burgy G, Guiheneuf S, Herault Y, Kendall DA, Carreaux F, Bazureau JP, Meijer L. Structure-Activity Relationship in the Leucettine Family of Kinase Inhibitors. J Med Chem 2021; 65:1396-1417. [PMID: 34928152 DOI: 10.1021/acs.jmedchem.1c01141] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The protein kinase DYRK1A is involved in Alzheimer's disease, Down syndrome, diabetes, viral infections, and leukemia. Leucettines, a family of 2-aminoimidazolin-4-ones derived from the marine sponge alkaloid Leucettamine B, have been developed as pharmacological inhibitors of DYRKs (dual specificity, tyrosine phosphorylation regulated kinases) and CLKs (cdc2-like kinases). We report here on the synthesis and structure-activity relationship (SAR) of 68 Leucettines. Leucettines were tested on 11 purified kinases and in 5 cellular assays: (1) CLK1 pre-mRNA splicing, (2) Threonine-212-Tau phosphorylation, (3) glutamate-induced cell death, (4) autophagy and (5) antagonism of ligand-activated cannabinoid receptor CB1. The Leucettine SAR observed for DYRK1A is essentially identical for CLK1, CLK4, DYRK1B, and DYRK2. DYRK3 and CLK3 are less sensitive to Leucettines. In contrast, the cellular SAR highlights correlations between inhibition of specific kinase targets and some but not all cellular effects. Leucettines deserve further development as potential therapeutics against various diseases on the basis of their molecular targets and cellular effects.
Collapse
Affiliation(s)
- Tania Tahtouh
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France.,CNRS, 'Protein Phosphorylation and Human Disease' Group, Station Biologique De Roscoff, Place G. Teissier, Bp 74, 29682 Roscoff, Bretagne, France.,College Of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | - Emilie Durieu
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France.,CNRS, 'Protein Phosphorylation and Human Disease' Group, Station Biologique De Roscoff, Place G. Teissier, Bp 74, 29682 Roscoff, Bretagne, France
| | - Benoît Villiers
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France
| | - Céline Bruyère
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France
| | - Thu Lan Nguyen
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France.,Institut De Génétique Et De Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, Université de Strasbourg, CNRS UMR7104 & INSERM U964, 67400 Illkirch, France.,Laboratory of Molecular & Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, New York 10021-6399, United States
| | - Xavier Fant
- CNRS, 'Protein Phosphorylation and Human Disease' Group, Station Biologique De Roscoff, Place G. Teissier, Bp 74, 29682 Roscoff, Bretagne, France
| | - Kwang H Ahn
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Storrs, Connecticut 06269, United States
| | - Leepakshi Khurana
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Storrs, Connecticut 06269, United States
| | - Emmanuel Deau
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France
| | - Mattias F Lindberg
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France
| | - Elodie Sévère
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France
| | - Frédéric Miege
- Edelris, Bâtiment Bioserra 1, 60 avenue Rockefeller, 69008 Lyon, France
| | - Didier Roche
- Edelris, Bâtiment Bioserra 1, 60 avenue Rockefeller, 69008 Lyon, France
| | - Emmanuelle Limanton
- Institut des Sciences Chimiques de Rennes ISCR-UMR CNRS 6226, Université de Rennes 1, Campus de Beaulieu, Bât. 10A, CS 74205, 263 Avenue du Général Leclerc, 35042 Rennes Cedex, France
| | - Jean-Martial L'Helgoual'ch
- Institut des Sciences Chimiques de Rennes ISCR-UMR CNRS 6226, Université de Rennes 1, Campus de Beaulieu, Bât. 10A, CS 74205, 263 Avenue du Général Leclerc, 35042 Rennes Cedex, France
| | - Guillaume Burgy
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France.,Institut des Sciences Chimiques de Rennes ISCR-UMR CNRS 6226, Université de Rennes 1, Campus de Beaulieu, Bât. 10A, CS 74205, 263 Avenue du Général Leclerc, 35042 Rennes Cedex, France
| | - Solène Guiheneuf
- Institut des Sciences Chimiques de Rennes ISCR-UMR CNRS 6226, Université de Rennes 1, Campus de Beaulieu, Bât. 10A, CS 74205, 263 Avenue du Général Leclerc, 35042 Rennes Cedex, France
| | - Yann Herault
- Institut De Génétique Et De Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, Université de Strasbourg, CNRS UMR7104 & INSERM U964, 67400 Illkirch, France
| | - Debra A Kendall
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Storrs, Connecticut 06269, United States
| | - François Carreaux
- Institut des Sciences Chimiques de Rennes ISCR-UMR CNRS 6226, Université de Rennes 1, Campus de Beaulieu, Bât. 10A, CS 74205, 263 Avenue du Général Leclerc, 35042 Rennes Cedex, France
| | - Jean-Pierre Bazureau
- Institut des Sciences Chimiques de Rennes ISCR-UMR CNRS 6226, Université de Rennes 1, Campus de Beaulieu, Bât. 10A, CS 74205, 263 Avenue du Général Leclerc, 35042 Rennes Cedex, France
| | - Laurent Meijer
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France
| |
Collapse
|
62
|
Brod SA. Anti-Inflammatory Agents: An Approach to Prevent Cognitive Decline in Alzheimer's Disease. J Alzheimers Dis 2021; 85:457-472. [PMID: 34842189 DOI: 10.3233/jad-215125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Systemic inflammation is an organism's response to an assault by the non-self. However, that inflammation may predispose humans to illnesses targeted to organs, including Alzheimer's disease (AD). Lesions in AD have pro-inflammatory cytokines and activated microglial/monocyte/macrophage cells. Up to this point, clinical trials using anti-amyloid monoclonal antibodies have not shown success. Maybe it is time to look elsewhere by combating inflammation. Neuroinflammation with CNS cellular activation and excessive expression of immune cytokines is suspected as the "principal culprit" in the higher risk for sporadic AD. Microglia, the resident immune cell of the CNS, perivascular myeloid cells, and activated macrophages produce IL-1, IL-6 at higher levels in patients with AD. Anti-inflammatory measures that target cellular/cytokine-mediated damage provide a rational therapeutic strategy. We propose a clinical trial using oral type 1 IFNs to act as such an agent; one that decreases IL-1 and IL-6 secretion by activating lamina propria lymphocytes in the gut associated lymphoid tissue with subsequent migration to the brain undergoing inflammatory responses. A clinical trial would be double-blind, parallel 1-year clinical trial randomized 1 : 1 oral active type 1 IFN versus best medical therapy to determine whether ingested type I IFN would decrease the rate of cognitive decline in mild cognitive impairment or mild AD. Using cognitive psychometrics, imaging, and fluid biomarkers (MxA for effective type I IFN activity beyond the gut), we can determine if oral type I IFN can prevent cognitive decline in AD.
Collapse
Affiliation(s)
- Staley A Brod
- Department of Neurology, Medical College of Wisconsin, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
63
|
Plasma phosphorylated-tau181 as a predictive biomarker for Alzheimer's amyloid, tau and FDG PET status. Transl Psychiatry 2021; 11:585. [PMID: 34775468 PMCID: PMC8590691 DOI: 10.1038/s41398-021-01709-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 10/17/2021] [Accepted: 10/26/2021] [Indexed: 12/13/2022] Open
Abstract
Plasma phosphorylated-tau181 (p-tau181) showed the potential for Alzheimer's diagnosis and prognosis, but its role in detecting cerebral pathologies is unclear. We aimed to evaluate whether it could serve as a marker for Alzheimer's pathology in the brain. A total of 1189 participants with plasma p-tau181 and PET data of amyloid, tau or FDG PET were included from ADNI. Cross-sectional relationships of plasma p-tau181 with PET biomarkers were tested. Longitudinally, we further investigated whether different p-tau181 levels at baseline predicted different progression of Alzheimer's pathological changes in the brain. We found plasma p-tau181 significantly correlated with brain amyloid (Spearman ρ = 0.45, P < 0.0001), tau (0.25, P = 0.0003), and FDG PET uptakes (-0.37, P < 0.0001), and increased along the Alzheimer's continuum. Individually, plasma p-tau181 could detect abnormal amyloid, tau pathologies and hypometabolism in the brain, similar with or even better than clinical indicators. The diagnostic accuracy of plasma p-tau181 elevated significantly when combined with clinical information (AUC = 0.814 for amyloid PET, 0.773 for tau PET, and 0.708 for FDG PET). Relationships of plasma p-tau181 with brain pathologies were partly or entirely mediated by the corresponding CSF biomarkers. Besides, individuals with abnormal plasma p-tau181 level (>18.85 pg/ml) at baseline had a higher risk of pathological progression in brain amyloid (HR: 2.32, 95%CI 1.32-4.08) and FDG PET (3.21, 95%CI 2.06-5.01) status. Plasma p-tau181 may be a sensitive screening test for detecting brain pathologies, and serve as a predictive biomarker for Alzheimer's pathophysiology.
Collapse
|
64
|
Altuna-Azkargorta M, Mendioroz-Iriarte M. Blood biomarkers in Alzheimer's disease. NEUROLOGÍA (ENGLISH EDITION) 2021; 36:704-710. [PMID: 34752348 DOI: 10.1016/j.nrleng.2018.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 03/01/2018] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Early diagnosis of Alzheimer disease (AD) through the use of biomarkers could assist in the implementation and monitoring of early therapeutic interventions, and has the potential to significantly modify the course of the disease. DEVELOPMENT The classic cerebrospinal fluid and approved structural and functional neuroimaging biomarkers are of limited clinical application given their invasive nature and/or high cost. The identification of more accessible and less costly biomarkers, such as blood biomarkers, would increase their use in clinical practice. We review the available published evidence on the main blood biochemical biomarkers potentially useful for diagnosing AD. CONCLUSIONS Blood biomarkers are more cost- and time-effective than CSF biomarkers. However, immediate applicability in clinical practice is relatively unlikely. The main limitations come from the difficulty of measuring and standardising thresholds between different laboratories and the failure to replicate results. Of all the molecules studied, apoptosis and neurodegeneration biomarkers and the biomarker panels obtained through "omics" approaches, such as isolated or combined metabolomics, offer the most promising results.
Collapse
Affiliation(s)
- M Altuna-Azkargorta
- Laboratorio de Neuroepigenética, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain.
| | - M Mendioroz-Iriarte
- Laboratorio de Neuroepigenética, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain; Servicio de Neurología, Complejo Hospitalario de Navarra, Pamplona, Spain
| |
Collapse
|
65
|
Singh K, Cheung BM, Xu A. Ultrasensitive detection of blood biomarkers of Alzheimer's and Parkinson's diseases: a systematic review. Biomark Med 2021; 15:1693-1708. [PMID: 34743546 DOI: 10.2217/bmm-2021-0219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose: Neurodegenerative disorders are a global health burden with costly and invasive diagnoses relying on brain imaging technology or CSF-based biomarkers. Therefore, considerable efforts to identify blood-biomarkers for Alzheimer's (AD) and Parkinson's diseases (PD) are ongoing. Objectives: This review evaluates the blood biomarkers for AD and PD for their diagnostic value. Methods: This study systematically reviewed articles published between July 1984 and February 2021. Among 1266 papers, we selected 42 studies for a systematic review and 23 studies for meta-analysis. Results & conclusion: Our analysis highlights P-tau181, T-tau and Nfl as promising blood biomarkers for AD diagnosis. Nfl levels were consistently raised in 16 AD and three PD cohorts. P-tau181 and T-tau were also significantly increased in 12 and eight AD cohorts, respectively.
Collapse
Affiliation(s)
- Kailash Singh
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| | - Bernard My Cheung
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China.,Department of Pharmacy & Pharmacology, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
66
|
Korecka M, Shaw LM. Mass spectrometry-based methods for robust measurement of Alzheimer's disease biomarkers in biological fluids. J Neurochem 2021; 159:211-233. [PMID: 34244999 PMCID: PMC9057379 DOI: 10.1111/jnc.15465] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/11/2021] [Accepted: 07/06/2021] [Indexed: 11/29/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia affecting 60%-70% of people afflicted with this disease. Accurate antemortem diagnosis is urgently needed for early detection of AD to enable reliable estimation of prognosis, intervention, and monitoring of the disease. The National Institute on Aging/Alzheimer's Association sponsored the 'Research Framework: towards a biological definition of AD', which recommends using different biomarkers in living persons for a biomarker-based definition of AD regardless of clinical status. Fluid biomarkers represent one of key groups of them. Since cerebrospinal fluid (CSF) is in direct contact with brain and many proteins present in the brain can be detected in CSF, this fluid has been regarded as the best biofluid in which to measure AD biomarkers. Recently, technological advancements in protein detection made possible the effective study of plasma AD biomarkers despite their significantly lower concentrations versus to that in CSF. This and other challenges that face plasma-based biomarker measurements can be overcome by using mass spectrometry. In this review, we discuss AD biomarkers which can be reliably measured in CSF and plasma using targeted mass spectrometry coupled to liquid chromatography (LC/MS/MS). We describe progress in LC/MS/MS methods' development, emphasize the challenges, and summarize major findings. We also highlight the role of mass spectrometry and progress made in the process of global standardization of the measurement of Aβ42/Aβ40. Finally, we briefly describe exploratory proteomics which seek to identify new biomarkers that can contribute to detection of co-pathological processes that are common in sporadic AD.
Collapse
Affiliation(s)
- Magdalena Korecka
- Department of Pathology and Laboratory Medicine Perlman School of Medicine University of Pennsylvania Philadelphia PA USA
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine Perlman School of Medicine University of Pennsylvania Philadelphia PA USA
| |
Collapse
|
67
|
Sugimoto T, Sakurai T, Akatsu H, Doi T, Fujiwara Y, Hirakawa A, Kinoshita F, Kuzuya M, Lee S, Matsuo K, Michikawa M, Ogawa S, Otsuka R, Sato K, Shimada H, Suzuki H, Suzuki H, Takechi H, Takeda S, Umegaki H, Wakayama S, Arai H. The Japan-Multimodal Intervention Trial for Prevention of Dementia (J-MINT): The Study Protocol for an 18-Month, Multicenter, Randomized, Controlled Trial. JPAD-JOURNAL OF PREVENTION OF ALZHEIMERS DISEASE 2021; 8:465-476. [PMID: 34585222 PMCID: PMC8187136 DOI: 10.14283/jpad.2021.29] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background/Objectives The Japan-multimodal intervention trial for prevention of dementia (J-MINT) is intended to verify the effectiveness of multi-domain interventions and to clarify the mechanism of cognitive improvement and deterioration by carrying out assessment of dementia-related biomarkers, omics analysis and brain imaging analysis among older adults at high risk of dementia. Moreover, the J-MINT trial collaborates with partnering private enterprises in the implementation of relevant interventional measures. This manuscript describes the study protocol. Design/Setting Eighteen-month, multi-centered, randomized controlled trial. Participants We plan to recruit 500 older adults aged 65–85 years with mild cognitive impairment. Subjects will be centrally randomized into intervention and control groups at a 1:1 allocation ratio using the dynamic allocation method with all subjects stratified by age, sex, and cognition. Intervention The multi-domain intervention program includes: (1) management of vascular risk factors; (2) group-based physical exercise and self-monitoring of physical activity; (3) nutritional counseling; and (4) cognitive training. Health-related information will be provided to the control group every two months. Measurements The primary and secondary outcomes will be assessed at baseline, 6-, 12-, and 18-month follow-up. The primary outcome is the change from baseline to 18 months in a global composite score combining several neuropsychological domains. Secondary outcomes include: cognitive change in each neuropsychological test, incident dementia, changes in blood and dementia-related biomarkers, changes in geriatric assessment including activities of daily living, frailty status and neuroimaging, and number of medications taken. Conclusions This trial that enlist the support of private enterprises will lead to the creation of new services for dementia prevention as well as to verify the effectiveness of multi-domain interventions for dementia prevention. Electronic Supplementary Material Supplementary material is available for this article at 10.14283/jpad.2021.29 and is accessible for authorized users.
Collapse
Affiliation(s)
- T Sugimoto
- Takashi Sakurai, 7-430 Morioka, Obu, Aichi, 474-8511, Japan, Tel: +81-562-46-2311, E-mail:
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Koychev I, Jansen K, Dette A, Shi L, Holling H. Blood-Based ATN Biomarkers of Alzheimer's Disease: A Meta-Analysis. J Alzheimers Dis 2021; 79:177-195. [PMID: 33252080 DOI: 10.3233/jad-200900] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The Amyloid Tau Neurodegeneration (ATN) framework was proposed to define the biological state underpinning Alzheimer's disease (AD). Blood-based biomarkers offer a scalable alternative to the costly and invasive currently available biomarkers. OBJECTIVE In this meta-analysis we sought to assess the diagnostic performance of plasma amyloid (Aβ40, Aβ42, Aβ42/40 ratio), tangle (p-tau181), and neurodegeneration (total tau [t-tau], neurofilament light [NfL]) biomarkers. METHODS Electronic databases were screened for studies reporting biomarker concentrations for AD and control cohorts. Biomarker performance was examined by random-effect meta-analyses based on the ratio between biomarker concentrations in patients and controls. RESULTS 83 studies published between 1996 and 2020 were included in the analyses. Aβ42/40 ratio as well as Aβ42 discriminated AD patients from controls when using novel platforms such as immunomagnetic reduction (IMR). We found significant differences in ptau-181 concentration for studies based on single molecule array (Simoa), but not for studies based on IMR or ELISA. T-tau was significantly different between AD patients and control in IMR and Simoa but not in ELISA-based studies. In contrast, NfL differentiated between groups across platforms. Exosome studies showed strong separation between patients and controls for Aβ42, t-tau, and p-tau181. CONCLUSION Currently available assays for sampling plasma ATN biomarkers appear to differentiate between AD patients and controls. Novel assay methodologies have given the field a significant boost for testing these biomarkers, such as IMR for Aβ, Simoa for p-tau181. Enriching samples through extracellular vesicles shows promise but requires further validation.
Collapse
Affiliation(s)
- Ivan Koychev
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Katrin Jansen
- Department of Psychology, University of Münster, Münster, Germany
| | - Alina Dette
- Department of Psychology, University of Münster, Münster, Germany
| | - Liu Shi
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Heinz Holling
- Department of Psychology, University of Münster, Münster, Germany
| |
Collapse
|
69
|
Cianflone A, Coppola L, Mirabelli P, Salvatore M. Predictive Accuracy of Blood-Derived Biomarkers for Amyloid-β Brain Deposition Along with the Alzheimer's Disease Continuum: A Systematic Review. J Alzheimers Dis 2021; 84:393-407. [PMID: 34542072 DOI: 10.3233/jad-210496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND An amyloid-β (Aβ) positron emission tomography (Aβ-PET) scan of the human brain could lead to an early diagnosis of Alzheimer's disease (AD) and estimate disease progression. However, Aβ-PET imaging is expensive, invasive, and rarely applicable to cognitively normal subjects at risk for dementia. The identification of blood biomarkers predictive of Aβ brain deposition could help the identification of subjects at risk for dementia and could be helpful for the prognosis of AD progression. OBJECTIVE This study aimed to analyze the prognostic accuracy of blood biomarkers in predicting Aβ-PET status along with progression toward AD. METHODS In accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we searched bibliographic databases from 2010 to 2020. The quality of the included studies was assessed by the Quality Assessment of Diagnostic Accuracy Studies 2 (QUADAS-2) tool. RESULTS A total of 8 studies were retrieved. The prognostic accuracy of Aβ-PET status was calculated by obtaining ROCs for the following biomarkers: free, total, and bound Aβ42 and Aβ40; Aβ42/40 ratio; neurofilaments (NFL); total tau (T-tau); and phosphorylated-tau181 (P-tau181). Higher and lower plasma baseline levels of P-tau181 and the Aβ42/40 ratio, respectively, showed consistently good prognostication of Aβ-PET brain accumulation. Only P-tau181 was shown to predict AD progression. CONCLUSION In conclusion, the Aβ42/40 ratio and plasma P-tau181 were shown to predict Aβ-PET status. Plasma P-tau181 could also be a preclinical biomarker for AD progression.
Collapse
|
70
|
Transferrin Biosynthesized in the Brain Is a Novel Biomarker for Alzheimer's Disease. Metabolites 2021; 11:metabo11090616. [PMID: 34564432 PMCID: PMC8470343 DOI: 10.3390/metabo11090616] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 01/23/2023] Open
Abstract
Glycosylation is a cell type-specific post-translational modification that can be used for biomarker identification in various diseases. Aim of this study is to explore glycan-biomarkers on transferrin (Tf) for Alzheimer’s disease (AD) in cerebrospinal fluid (CSF). Glycan structures of CSF Tf were analyzed by ultra-performance liquid chromatography followed by mass spectrometry. We found that a unique mannosylated-glycan is carried by a Tf isoform in CSF (Man-Tf). The cerebral cortex contained Man-Tf as a major isofom, suggesting that CSF Man-Tf is, at least partly, derived from the cortex. Man-Tf levels were analyzed in CSF of patients with neurological diseases. Concentrations of Man-Tf were significantly increased in AD and mild cognitive impairment (MCI) comparing with other neurological diseases, and the levels correlated well with those of phosphorylated-tau (p-tau), a representative AD marker. Consistent with the observation, p-tau and Tf were co-expressed in hippocampal neurons of AD, leading to the notion that a combined p-tau and Man-Tf measure could be a biomarker for AD. Indeed, levels of p-tau x Man-Tf showed high diagnostic accuracy for MCI and AD; 84% sensitivities and 90% specificities for MCI and 94% sensitivities and 89% specificities for AD. Thus Man-Tf could be a new biomarker for AD.
Collapse
|
71
|
Alawode DOT, Heslegrave AJ, Ashton NJ, Karikari TK, Simrén J, Montoliu‐Gaya L, Pannee J, O´Connor A, Weston PSJ, Lantero‐Rodriguez J, Keshavan A, Snellman A, Gobom J, Paterson RW, Schott JM, Blennow K, Fox NC, Zetterberg H. Transitioning from cerebrospinal fluid to blood tests to facilitate diagnosis and disease monitoring in Alzheimer's disease. J Intern Med 2021; 290:583-601. [PMID: 34021943 PMCID: PMC8416781 DOI: 10.1111/joim.13332] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/18/2021] [Accepted: 03/30/2021] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is increasingly prevalent worldwide, and disease-modifying treatments may soon be at hand; hence, now, more than ever, there is a need to develop techniques that allow earlier and more secure diagnosis. Current biomarker-based guidelines for AD diagnosis, which have replaced the historical symptom-based guidelines, rely heavily on neuroimaging and cerebrospinal fluid (CSF) sampling. While these have greatly improved the diagnostic accuracy of AD pathophysiology, they are less practical for application in primary care, population-based and epidemiological settings, or where resources are limited. In contrast, blood is a more accessible and cost-effective source of biomarkers in AD. In this review paper, using the recently proposed amyloid, tau and neurodegeneration [AT(N)] criteria as a framework towards a biological definition of AD, we discuss recent advances in biofluid-based biomarkers, with a particular emphasis on those with potential to be translated into blood-based biomarkers. We provide an overview of the research conducted both in CSF and in blood to draw conclusions on biomarkers that show promise. Given the evidence collated in this review, plasma neurofilament light chain (N) and phosphorylated tau (p-tau; T) show particular potential for translation into clinical practice. However, p-tau requires more comparisons to be conducted between its various epitopes before conclusions can be made as to which one most robustly differentiates AD from non-AD dementias. Plasma amyloid beta (A) would prove invaluable as an early screening modality, but it requires very precise tests and robust pre-analytical protocols.
Collapse
Affiliation(s)
- D. O. T. Alawode
- From theDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - A. J. Heslegrave
- From theDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - N. J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational MedicineDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Department of Old Age PsychiatryInstitute of Psychiatry, Psychology & NeuroscienceKing’s College LondonLondonUK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS FoundationLondonUK
| | - T. K. Karikari
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - J. Simrén
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - L. Montoliu‐Gaya
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - J. Pannee
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - A. O´Connor
- UK Dementia Research Institute at UCLLondonUK
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - P. S. J. Weston
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - J. Lantero‐Rodriguez
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - A. Keshavan
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - A. Snellman
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Turku PET CentreUniversity of TurkuTurkuFinland
| | - J. Gobom
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - R. W. Paterson
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - J. M. Schott
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - K. Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - N. C. Fox
- UK Dementia Research Institute at UCLLondonUK
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - H. Zetterberg
- From theDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| |
Collapse
|
72
|
Muraoka S, DeLeo AM, Yang Z, Tatebe H, Yukawa-Takamatsu K, Ikezu S, Tokuda T, Issadore D, Stern RA, Ikezu T. Proteomic Profiling of Extracellular Vesicles Separated from Plasma of Former National Football League Players at Risk for Chronic Traumatic Encephalopathy. Aging Dis 2021; 12:1363-1375. [PMID: 34527415 PMCID: PMC8407879 DOI: 10.14336/ad.2020.0908] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022] Open
Abstract
Chronic Traumatic Encephalopathy (CTE) is a tauopathy that affects individuals with a history of exposure to repetitive head impacts, including National Football League (NFL) players. Extracellular vesicles (EVs) are known to carry tau in Alzheimer's disease and other tauopathies. We examined protein profiles of EVs separated from the plasma of former NFL players at risk for CTE. EVs were separated from the plasma from former NFL players and age-matched controls using size-exclusion chromatography. Label-free quantitative proteomic analysis identified 675 proteins in plasma EVs, and 17 proteins were significantly differentially expressed between former NFL players and controls. Total tau (t-tau) and tau phosphorylated at threonie181 (p-tau181) in plasma-derived EVs were measured by ultrasensitive immunoassay. Level of t-tau and p-tau181 in EVs were significantly different, and the area under the receiver operating characteristic curve (AUC) of t-tau and p-tau181 showed 0.736 and 0.715, respectively. Machine learning analysis indicated that a combination of collagen type VI alpha 3 and 1 chain (COL6A3 and COL6A1) and reelin (RELN) can distinguish former NFL players from controls with 85% accuracy (AUC = 0.85). Based on the plasma EV proteomics, these data provide protein profiling of plasma EVs for CTE, and indicate combination of COL6A3, RELN and COL6A1 in plasma EVs may serve as the potential diagnostic biomarkers for CTE.
Collapse
Affiliation(s)
- Satoshi Muraoka
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| | - Annina M DeLeo
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| | - Zijian Yang
- Deprtment of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
| | - Harutsugu Tatebe
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, JAPAN.
| | - Kayo Yukawa-Takamatsu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| | - Seiko Ikezu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| | - Takahiko Tokuda
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, JAPAN.
| | - David Issadore
- Deprtment of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
| | - Robert A Stern
- Department of Neurology, Boston University Alzheimer’s Disease and CTE Centers, Boston University School of Medicine, Boston, MA, USA.
- Departments of Anatomy & Neurobiology and Neurosurgery, Boston University School of Medicine, Boston, MA, USA.
- Center for Systems Neuroscience, Boston University, Boston, MA, USA.
| | - Tsuneya Ikezu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
- Department of Neurology, Boston University Alzheimer’s Disease and CTE Centers, Boston University School of Medicine, Boston, MA, USA.
- Center for Systems Neuroscience, Boston University, Boston, MA, USA.
| |
Collapse
|
73
|
Montoliu-Gaya L, Strydom A, Blennow K, Zetterberg H, Ashton NJ. Blood Biomarkers for Alzheimer's Disease in Down Syndrome. J Clin Med 2021; 10:3639. [PMID: 34441934 PMCID: PMC8397053 DOI: 10.3390/jcm10163639] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/09/2021] [Accepted: 08/12/2021] [Indexed: 12/15/2022] Open
Abstract
Epidemiological evidence suggests that by the age of 40 years, all individuals with Down syndrome (DS) have Alzheimer's disease (AD) neuropathology. Clinical diagnosis of dementia by cognitive assessment is complex in these patients due to the pre-existing and varying intellectual disability, which may mask subtle declines in cognitive functioning. Cerebrospinal fluid (CSF) and positron emission tomography (PET) biomarkers, although accurate, are expensive, invasive, and particularly challenging in such a vulnerable population. The advances in ultra-sensitive detection methods have highlighted blood biomarkers as a valuable and realistic tool for AD diagnosis. Studies with DS patients have proven the potential blood-based biomarkers for sporadic AD (amyloid-β, tau, phosphorylated tau, and neurofilament light chain) to be useful in this population. In addition, biomarkers related to other pathologies that could aggravate dementia progression-such as inflammatory dysregulation, energetic imbalance, or oxidative stress-have been explored. This review serves to provide a brief overview of the main findings from the limited neuroimaging and CSF studies, outline the current state of blood biomarkers to diagnose AD in patients with DS, discuss possible past limitations of the research, and suggest considerations for developing and validating blood-based biomarkers in the future.
Collapse
Affiliation(s)
- Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, 431 41 Mölndal, Sweden; (K.B.); (H.Z.); (N.J.A.)
| | - Andre Strydom
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London WC2R 2LS, UK;
- South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- London Down Syndrome Consortium (LonDowns), London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, 431 41 Mölndal, Sweden; (K.B.); (H.Z.); (N.J.A.)
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, 431 41 Mölndal, Sweden; (K.B.); (H.Z.); (N.J.A.)
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Nicholas James Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, 431 41 Mölndal, Sweden; (K.B.); (H.Z.); (N.J.A.)
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London SE5 9RT, UK
- NIHR Biomedical Research Centre for Mental Health, Biomedical Research Unit for Dementia at South London, Maudsley NHS Foundation, London SE5 8AF, UK
| |
Collapse
|
74
|
Shinohara M, Hirokawa J, Shimodaira A, Tashiro Y, Suzuki K, Gheni G, Fukumori A, Matsubara T, Morishima M, Saito Y, Murayama S, Sato N. ELISA Evaluation of Tau Accumulation in the Brains of Patients with Alzheimer Disease. J Neuropathol Exp Neurol 2021; 80:652-662. [PMID: 34283221 DOI: 10.1093/jnen/nlab047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Despite the routine use of sandwich enzyme-linked immunosorbent assays (ELISAs) for quantifying tau levels in CSF and plasma, tau accumulations in the brains of patients with Alzheimer disease (AD) have rarely been evaluated by this method. Thus, by introducing several tau ELISAs that target different epitopes, we evaluated accumulated tau levels in postmortem brains depending on disease stage, brain areas, and other AD-related changes. Notably, tau levels in insoluble fraction determined by each ELISAs differ depending on the epitopes of antibodies: non-AD control samples yield relatively high signals when an antibody against the N-terminal region of tau is used. On the other hand, ELISAs combining antibodies against the later-middle to C-terminal regions of tau produced substantially increased signals from AD samples, compared to those from non-AD controls. Such ELISAs better distinguish AD and non-AD controls, and the results are more closely associated with Braak neurofibrillary tangles stage, Aβ accumulation, and glial markers. Moreover, these ELISAs can reflect the pattern of tau spread across brain regions. In conclusion, Tau ELISAs that combine antibodies against the later-middle to C-terminal regions of tau can better reflect neuropathological tau accumulation, which would enable to evaluate tau accumulation in the brain at a biochemical level.
Collapse
Affiliation(s)
- Mitsuru Shinohara
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Junko Hirokawa
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Akemi Shimodaira
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Yoshitaka Tashiro
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Kaoru Suzuki
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Ghupurjan Gheni
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Akio Fukumori
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Tomoyasu Matsubara
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Maho Morishima
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Yuko Saito
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Shigeo Murayama
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Naoyuki Sato
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| |
Collapse
|
75
|
Phosphorylated tau181 in plasma as a potential biomarker for Alzheimer's disease in adults with Down syndrome. Nat Commun 2021; 12:4304. [PMID: 34262030 PMCID: PMC8280160 DOI: 10.1038/s41467-021-24319-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 06/09/2021] [Indexed: 12/14/2022] Open
Abstract
Plasma tau phosphorylated at threonine 181 (p-tau181) predicts Alzheimer’s disease (AD) pathology with high accuracy in the general population. In this study, we investigated plasma p-tau181 as a biomarker of AD in individuals with Down syndrome (DS). We included 366 adults with DS (240 asymptomatic, 43 prodromal AD, 83 AD dementia) and 44 euploid cognitively normal controls. We measured plasma p-tau181 with a Single molecule array (Simoa) assay. We examined the diagnostic performance of p-tau181 for the detection of AD and the relationship with other fluid and imaging biomarkers. Plasma p-tau181 concentration showed an area under the curve of 0.80 [95% CI 0.73–0.87] and 0.92 [95% CI 0.89–0.95] for the discrimination between asymptomatic individuals versus those in the prodromal and dementia groups, respectively. Plasma p-tau181 correlated with atrophy and hypometabolism in temporoparietal regions. Our findings indicate that plasma p-tau181 concentration can be useful to detect AD in DS. Plasma tau phosphorylated at threonine 181 (p-tau181) predicts Alzheimer’s disease (AD) pathology. Here, the authors investigated whether plasma ptau181 could be a potential biomarker of AD in individuals with Down syndrome (DS) and find plasma p-tau181 can detect AD in DS adults.
Collapse
|
76
|
Ashton NJ, Leuzy A, Karikari TK, Mattsson-Carlgren N, Dodich A, Boccardi M, Corre J, Drzezga A, Nordberg A, Ossenkoppele R, Zetterberg H, Blennow K, Frisoni GB, Garibotto V, Hansson O. The validation status of blood biomarkers of amyloid and phospho-tau assessed with the 5-phase development framework for AD biomarkers. Eur J Nucl Med Mol Imaging 2021; 48:2140-2156. [PMID: 33677733 PMCID: PMC8175325 DOI: 10.1007/s00259-021-05253-y] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/09/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE The development of blood biomarkers that reflect Alzheimer's disease (AD) pathophysiology (phosphorylated tau and amyloid-β) has offered potential as scalable tests for dementia differential diagnosis and early detection. In 2019, the Geneva AD Biomarker Roadmap Initiative included blood biomarkers in the systematic validation of AD biomarkers. METHODS A panel of experts convened in November 2019 at a two-day workshop in Geneva. The level of maturity (fully achieved, partly achieved, preliminary evidence, not achieved, unsuccessful) of blood biomarkers was assessed based on the Biomarker Roadmap methodology and discussed fully during the workshop which also evaluated cerebrospinal fluid (CSF) and positron emission tomography (PET) biomarkers. RESULTS Plasma p-tau has shown analytical validity (phase 2 primary aim 1) and first evidence of clinical validity (phase 3 primary aim 1), whereas the maturity level for Aβ remains to be partially achieved. Full and partial achievement has been assigned to p-tau and Aβ, respectively, in their associations to ante-mortem measures (phase 2 secondary aim 2). However, only preliminary evidence exists for the influence of covariates, assay comparison and cut-off criteria. CONCLUSIONS Despite the relative infancy of blood biomarkers, in comparison to CSF biomarkers, much has already been achieved for phases 1 through 3 - with p-tau having greater success in detecting AD and predicting disease progression. However, sufficient data about the effect of covariates on the biomarker measurement is lacking. No phase 4 (real-world performance) or phase 5 (assessment of impact/cost) aim has been tested, thus not achieved.
Collapse
Affiliation(s)
- N J Ashton
- Institute of Neuroscience & Physiology, Department of Psychiatry & Neurochemistry, Sahlgrenska Academy, University of Gothenburg, House V3/SU, SE-431 80, Mölndal, Sweden.
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - A Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - T K Karikari
- Institute of Neuroscience & Physiology, Department of Psychiatry & Neurochemistry, Sahlgrenska Academy, University of Gothenburg, House V3/SU, SE-431 80, Mölndal, Sweden
| | - N Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - A Dodich
- NIMTlab - Neuroimaging and Innovative Molecular Tracers Laboratory, University of Geneva, Geneva, Switzerland
- Center for Neurocognitive Rehabilitation (CeRiN), CIMeC, University of Trento, Trento, Italy
| | - M Boccardi
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald, Rostock, Germany
- LANVIE - Laboratory of Neuroimaging of Aging, University of Geneva, Geneva, Switzerland
| | - J Corre
- Centre National de la Recherche Scientifique, Montpellier, France
| | - A Drzezga
- Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - A Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Aging, Karolinska University Hospital Stockholm, Stockholm, Sweden
| | - R Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - H Zetterberg
- Institute of Neuroscience & Physiology, Department of Psychiatry & Neurochemistry, Sahlgrenska Academy, University of Gothenburg, House V3/SU, SE-431 80, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - K Blennow
- Institute of Neuroscience & Physiology, Department of Psychiatry & Neurochemistry, Sahlgrenska Academy, University of Gothenburg, House V3/SU, SE-431 80, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - G B Frisoni
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald, Rostock, Germany
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
| | - V Garibotto
- NIMTlab - Neuroimaging and Innovative Molecular Tracers Laboratory, University of Geneva, Geneva, Switzerland
- Diagnostic Department, University Hospitals of Geneva, Geneva, Switzerland
| | - O Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.
- UK Dementia Research Institute at UCL, London, UK.
- Memory Clinic, Skåne University Hospital, SE-205 02, Malmö, Sweden.
| |
Collapse
|
77
|
Kondo T, Banno H, Okunomiya T, Amino Y, Endo K, Nakakura A, Uozumi R, Kinoshita A, Tada H, Morita S, Ishikawa H, Shindo A, Yasuda K, Taruno Y, Maki T, Suehiro T, Mori K, Ikeda M, Fujita K, Izumi Y, Kanemaru K, Ishii K, Shigenobu K, Kutoku Y, Sunada Y, Kawakatsu S, Shiota S, Watanabe T, Uchikawa O, Takahashi R, Tomimoto H, Inoue H. Repurposing bromocriptine for Aβ metabolism in Alzheimer's disease (REBRAnD) study: randomised placebo-controlled double-blind comparative trial and open-label extension trial to investigate the safety and efficacy of bromocriptine in Alzheimer's disease with presenilin 1 (PSEN1) mutations. BMJ Open 2021; 11:e051343. [PMID: 34193504 PMCID: PMC8246358 DOI: 10.1136/bmjopen-2021-051343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
INTRODUCTION Alzheimer's disease (AD) is one of the most common causes of dementia. Pathogenic variants in the presenilin 1 (PSEN1) gene are the most frequent cause of early-onset AD. Medications for patients with AD bearing PSEN1 mutation (PSEN1-AD) are limited to symptomatic therapies and no established radical treatments are available. Induced pluripotent stem cell (iPSC)-based drug repurposing identified bromocriptine as a therapeutic candidate for PSEN1-AD. In this study, we used an enrichment strategy with iPSCs to select the study population, and we will investigate the safety and efficacy of an orally administered dose of bromocriptine in patients with PSEN1-AD. METHODS AND ANALYSIS This is a multicentre, randomised, placebo-controlled trial. AD patients with PSEN1 mutations and a Mini Mental State Examination-Japanese score of ≤25 will be randomly assigned, at a 2:1 ratio, to the trial drug or placebo group (≥4 patients in TW-012R and ≥2 patients in placebo). This clinical trial consists of a screening period, double-blind phase (9 months) and extension phase (3 months). The double-blind phase for evaluating the efficacy and safety is composed of the low-dose maintenance period (10 mg/day), high-dose maintenance period (22.5 mg/day) and tapering period of the trial drug. Additionally, there is an open-labelled active drug extension period for evaluating long-term safety. Primary outcomes are safety and efficacy in cognitive and psychological function. Also, exploratory investigations for the efficacy of bromocriptine by neurological scores and biomarkers will be conducted. ETHICS AND DISSEMINATION The proposed trial is conducted according to the Declaration of Helsinki, and was approved by the Institutional Review Board (K070). The study results are expected to be disseminated at international or national conferences and published in international journals following the peer-review process. TRIAL REGISTRATION NUMBER jRCT2041200008, NCT04413344.
Collapse
Affiliation(s)
- Takayuki Kondo
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Haruhiko Banno
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Taro Okunomiya
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Yoko Amino
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Kayoko Endo
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Akiyoshi Nakakura
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Ryuji Uozumi
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akemi Kinoshita
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Harue Tada
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hidehiro Ishikawa
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Ken Yasuda
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yosuke Taruno
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takakuni Maki
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takashi Suehiro
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kohji Mori
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Koji Fujita
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yuishin Izumi
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kazutomi Kanemaru
- Department of Stroke, Tokyo Metropolitan Geriatric Medical Center, Tokyo, Japan
| | - Kenji Ishii
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | | | - Yumiko Kutoku
- Department of Neurology, Kawasaki Medical School, Kurashiki, Japan
| | - Yoshihide Sunada
- Department of Neurology, Kawasaki Medical School, Kurashiki, Japan
| | - Shinobu Kawakatsu
- Department of Neuropsychiatry, Fukushima Medical University Aizu Medical Center, Aizu, Japan
| | | | | | | | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
78
|
Winder Z, Wilcock D, Jicha GA. Diagnostic and Prognostic Laboratory Testing for Alzheimer Disease. Clin Lab Med 2021; 40:289-303. [PMID: 32718500 DOI: 10.1016/j.cll.2020.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This article focuses on current clinical laboratory testing to diagnose Alzheimer disease and monitor its progression throughout its disease course. Several clinically available tests focus on analysis of amyloid and tau levels in cerebrospinal fluid as well as autosomal dominant and risk factor genes. Although the current armament of clinical laboratory testing is limited by invasiveness of cerebrospinal fluid collection, rarity of autosomal dominant genetic mutations, and uncertainties of risk inherent in nonpenetrant genes, the field is poised to advance the clinical repertoire of laboratory diagnostic testing.
Collapse
Affiliation(s)
- Zachary Winder
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky College of Medicine, 800 South Limestone Street, Lexington, KY 40536-0230, USA
| | - Donna Wilcock
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky College of Medicine, 800 South Limestone Street, Lexington, KY 40536-0230, USA
| | - Gregory A Jicha
- Department of Neurology, Sanders-Brown Center on Aging, University of Kentucky College of Medicine, 800 South Limestone Street, Lexington, KY 40536-0230, USA.
| |
Collapse
|
79
|
Longitudinal plasma phosphorylated tau 181 tracks disease progression in Alzheimer's disease. Transl Psychiatry 2021; 11:356. [PMID: 34120152 PMCID: PMC8197760 DOI: 10.1038/s41398-021-01476-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/19/2021] [Accepted: 05/27/2021] [Indexed: 11/08/2022] Open
Abstract
To assess plasma phosphorylated tau181 (p-tau181) as a progression biomarker in Alzheimer's disease (AD), we examined longitudinal plasma p-tau181 of 1184 participants (403 cognitively normal (CN), 560 patients with mild cognitive impairment (MCI), and 221 with AD dementia) from Alzheimer's Disease Neuroimaging Initiative (ADNI). The plasma p-tau level was increased at baseline for MCI and AD dementia (mean: CN, 15.4 pg/mL; MCI, 18.4 pg/mL; AD dementia, 23.7 pg/mL; P < 0.001) and increased significantly over time at preclinical (Aβ-positive CN), prodromal (Aβ-positive MCI), and dementia (Aβ-positive dementia) stage of AD. A longitudinal increase of plasma p-tau181 was associated with abnormal cerebrospinal fluid biomarker levels (low Aβ42, high phosphorylated tau, and high total tau, all P < 0.001), amyloid accumulation (P < 0.001) and hypometabolism (P = 0.002) on positron emission tomography, atrophy in structure imaging (small hippocampal (P = 0.030), middle temporal (P = 0.008), and whole brain (P = 0.027) volume, and large ventricular volume (P = 0.008)), and deteriorated cognitive performance (global cognition and memory, language, executive function, and visuospatial function, all P < 0.050) at baseline. Furthermore, longitudinal plasma p-tau181 correlated with concurrent changes of nearly all these AD-related hallmarks and faster increase in plasma p-tau181 correlated with faster worsening cognition in all diagnostic groups. Importantly, most associations remained significant in Aβ-positive group and became non-significant in Aβ-negative group. Longitudinal analyses of plasma p-tau181 suggest its potential as a noninvasive biomarker to track disease progression in AD and to monitor effects of disease-modifying therapeutics in clinical trials.
Collapse
|
80
|
Nakai T, Yamada K, Mizoguchi H. Alzheimer's Disease Animal Models: Elucidation of Biomarkers and Therapeutic Approaches for Cognitive Impairment. Int J Mol Sci 2021; 22:5549. [PMID: 34074018 PMCID: PMC8197360 DOI: 10.3390/ijms22115549] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is an age-related and progressive neurodegenerative disorder. It is widely accepted that AD is mainly caused by the accumulation of extracellular amyloid β (Aβ) and intracellular neurofibrillary tau tangles. Aβ begins to accumulate years before the onset of cognitive impairment, suggesting that the benefit of currently available interventions would be greater if they were initiated in the early phases of AD. To understand the mechanisms of AD pathogenesis, various transgenic mouse models with an accelerated accumulation of Aβ and tau tangles have been developed. However, none of these models exhibit all pathologies present in human AD. To overcome these undesirable phenotypes, APP knock-in mice, which were presented with touchscreen-based tasks, were developed to better evaluate the efficacy of candidate therapeutics in mouse models of early-stage AD. This review assesses several AD mouse models from the aspect of biomarkers and cognitive impairment and discusses their potential as tools to provide novel AD therapeutic approaches.
Collapse
Affiliation(s)
- Tsuyoshi Nakai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan; (T.N.); (K.Y.)
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan; (T.N.); (K.Y.)
| | - Hiroyuki Mizoguchi
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan; (T.N.); (K.Y.)
- Medical Interactive Research and Academia Industry Collaboration Center, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| |
Collapse
|
81
|
Marcucci V, Kleiman J. Biomarkers and Their Implications in Alzheimer’s Disease: A Literature Review. EXPLORATORY RESEARCH AND HYPOTHESIS IN MEDICINE 2021; 000:000-000. [DOI: 10.14218/erhm.2021.00016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
82
|
Dong Y, Liang F, Huang L, Fang F, Yang G, Tanzi RE, Zhang Y, Quan Q, Xie Z. The anesthetic sevoflurane induces tau trafficking from neurons to microglia. Commun Biol 2021; 4:560. [PMID: 33980987 PMCID: PMC8115254 DOI: 10.1038/s42003-021-02047-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 03/29/2021] [Indexed: 01/08/2023] Open
Abstract
Accumulation and spread of tau in Alzheimer's disease and other tauopathies occur in a prion-like manner. However, the mechanisms and downstream consequences of tau trafficking remain largely unknown. We hypothesized that tau traffics from neurons to microglia via extracellular vesicles (EVs), leading to IL-6 generation and cognitive impairment. We assessed mice and neurons treated with anesthetics sevoflurane and desflurane, and applied nanobeam-sensor technology, an ultrasensitive method, to measure tau/p-tau amounts. Sevoflurane, but not desflurane, increased tau or p-tau amounts in blood, neuron culture medium, or EVs. Sevoflurane increased p-tau amounts in brain interstitial fluid. Microglia from tau knockout mice took up tau and p-tau when treated with sevoflurane-conditioned neuron culture medium, leading to IL-6 generation. Tau phosphorylation inhibitor lithium and EVs generation inhibitor GW4869 attenuated tau trafficking. GW4869 mitigated sevoflurane-induced cognitive impairment in mice. Thus, tau trafficking could occur from neurons to microglia to generate IL-6, leading to cognitive impairment.
Collapse
Affiliation(s)
- Yuanlin Dong
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Feng Liang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Lining Huang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Department of Anesthesiology, the Second Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Fang Fang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Guang Yang
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Yiying Zhang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Qimin Quan
- Rowland Institute at Harvard University, Cambridge, MA, USA
- NanoMosaic, Woburn, MA, USA
| | - Zhongcong Xie
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
83
|
Chen M, Xia W. Proteomic Profiling of Plasma and Brain Tissue from Alzheimer's Disease Patients Reveals Candidate Network of Plasma Biomarkers. J Alzheimers Dis 2021; 76:349-368. [PMID: 32474469 DOI: 10.3233/jad-200110] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most prevalent form of dementia with two pathological hallmarks of tau-containing neurofibrillary tangles and amyloid-β protein (Aβ)-containing neuritic plaques. Although Aβ and tau have been explored as potential biomarkers, levels of these pathological proteins in blood fail to distinguish AD from healthy control subjects. OBJECTIVE We aim to discover potential plasma proteins associated with AD pathology by performing tandem mass tag (TMT)-based quantitative proteomic analysis of proteins from peripheral and central nervous system compartments. METHODS We performed comparative proteomic analyses of plasma collected from AD patients and cognitively normal subjects. In addition, proteomic profiles from the inferior frontal cortex, superior frontal cortex, and cerebellum of postmortem brain tissue from five AD patients and five non-AD controls were compared with plasma proteomic profiles to search for common biomarkers. Liquid chromatography-mass spectrometry was used to analyze plasma and brain tissue labeled with isobaric TMT for relative protein quantification. RESULTS Our results showed that the proteins in complement coagulation cascade and interleukin-6 signaling were significantly altered in both plasma and brains of AD patients. CONCLUSION Our results demonstrate the relevance in immune responses between the peripheral and central nervous systems. Those differentially regulated plasma proteins are explored as candidate biomarker profiles that illustrate chronic neuroinflammation in brains of AD patients.
Collapse
Affiliation(s)
- Mei Chen
- Geriatric Research Education Clinical Center, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, USA
| | - Weiming Xia
- Geriatric Research Education Clinical Center, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, USA.,Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
84
|
Budelier MM, Bateman RJ. Biomarkers of Alzheimer Disease. J Appl Lab Med 2021; 5:194-208. [PMID: 31843944 DOI: 10.1373/jalm.2019.030080] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/31/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND Alzheimer disease (AD) was once a clinical diagnosis confirmed by postmortem autopsy. Today, with the development of AD biomarkers, laboratory assays to detect AD pathology are able to complement clinical diagnosis in symptomatic individuals with uncertain diagnosis. A variety of commercially available assays are performed as laboratory-developed tests, and many more are in development for both clinical and research purposes. CONTENT The role of laboratory medicine in diagnosing and managing AD is expanding; thus, it is important for laboratory professionals and ordering physicians to understand the strengths and limitations of both existing and emerging AD biomarker assays. In this review, we will provide an overview of the diagnosis of AD, discuss existing laboratory assays for AD and their recommended use, and examine the clinical performance of emerging AD biomarkers. SUMMARY The field of AD biomarker discovery and assay development is rapidly evolving, with recent studies promising to improve both the diagnosis of symptomatic individuals and enrollment and monitoring of asymptomatic individuals in research studies. However, care must be taken to ensure proper use and interpretation of these assays. For clinical purposes, these assays are meant to aid in diagnosis but are not themselves diagnostic. For individuals without symptoms, AD biomarker tests are still only appropriate for research purposes. Additionally, there are analytical challenges that require careful attention, especially for longitudinal use of AD tests.
Collapse
Affiliation(s)
- Melissa M Budelier
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
85
|
Mielke MM. Consideration of Sex Differences in the Measurement and Interpretation of Alzheimer Disease-Related Biofluid-Based Biomarkers. J Appl Lab Med 2021; 5:158-169. [PMID: 31811073 DOI: 10.1373/jalm.2019.030023] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/23/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND The development of cerebrospinal fluid and blood-based biomarkers for Alzheimer disease (AD) and related disorders is rapidly progressing. Such biomarkers may be used clinically to screen the population, to enhance diagnosis, or to help determine prognosis. Although the use of precision medicine methods has contributed to enhanced understanding of the AD pathophysiological changes and development of assays, one aspect not commonly considered is sex differences. CONTENT There are several ways in which sex can affect the concentration or interpretation of biofluid biomarkers. For some markers, concentrations will vary by sex. For others, the concentrations might not vary by sex, but the impact or interpretation may vary by sex depending on the context of use (e.g., diagnostic vs prognostic). Finally, for others, there will be no sex differences in concentrations or their interpretation. This review will first provide a basis for sex differences, including differences in brain structure and function, and the means by which these differences could contribute to sex differences in biomarker concentrations. Next, the current state of sex differences in AD-related biofluid markers (i.e., amyloid-β, phosphorylated τ, total τ, neurofilament light chain, and neurogranin) will be reviewed. Lastly, factors that can lead to the misinterpretation of observed sex differences in biomarkers (either providing evidence for or against) will be considered. SUMMARY This review is intended to provide an impetus to consider sex differences in the measurement and interpretation of AD-related biofluid-based biomarkers.
Collapse
Affiliation(s)
- Michelle M Mielke
- Departments of Health Sciences Research and Neurology, Mayo Clinic, Rochester, MN
| |
Collapse
|
86
|
Solje E, Benussi A, Buratti E, Remes AM, Haapasalo A, Borroni B. State-of-the-Art Methods and Emerging Fluid Biomarkers in the Diagnostics of Dementia-A Short Review and Diagnostic Algorithm. Diagnostics (Basel) 2021; 11:diagnostics11050788. [PMID: 33925655 PMCID: PMC8145467 DOI: 10.3390/diagnostics11050788] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022] Open
Abstract
The most common neurodegenerative dementias include Alzheimer’s disease (AD), dementia with Lewy bodies (DLB), and frontotemporal dementia (FTD). The correct etiology-based diagnosis is pivotal for clinical management of these diseases as well as for the suitable timing and choosing the accurate disease-modifying therapies when these become available. Enzyme-linked immunosorbent assay (ELISA)-based methods, detecting altered levels of cerebrospinal fluid (CSF) Tau, phosphorylated Tau, and Aβ-42 in AD, allowed the wide use of this set of biomarkers in clinical practice. These analyses demonstrate a high diagnostic accuracy in AD but suffer from a relatively restricted usefulness due to invasiveness and lack of prognostic value. In recent years, the development of novel advanced techniques has offered new state-of-the-art opportunities in biomarker discovery. These include single molecule array technology (SIMOA), a tool for non-invasive analysis of ultra-low levels of central nervous system-derived molecules from biofluids, such as CSF or blood, and real-time quaking (RT-QuIC), developed to analyze misfolded proteins. In the present review, we describe the history of methods used in the fluid biomarker analyses of dementia, discuss specific emerging biomarkers with translational potential for clinical use, and suggest an algorithm for the use of new non-invasive blood biomarkers in clinical practice.
Collapse
Affiliation(s)
- Eino Solje
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, 70211 Kuopio, Finland;
- Neuro Center, Neurology, Kuopio University Hospital, 70029 Kuopio, Finland
| | - Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, 25121 Brescia, Italy;
| | - Emanuele Buratti
- International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy;
| | - Anne M. Remes
- Research Unit of Clinical Neuroscience, Neurology, University of Oulu, 90230 Oulu, Finland;
- Medical Research Center (MRC), Oulu University Hospital, 90220 Oulu, Finland
| | - Annakaisa Haapasalo
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland;
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, 25121 Brescia, Italy;
- Correspondence:
| |
Collapse
|
87
|
Abe K, Shang J, Shi X, Yamashita T, Hishikawa N, Takemoto M, Morihara R, Nakano Y, Ohta Y, Deguchi K, Ikeda M, Ikeda Y, Okamoto K, Shoji M, Takatama M, Kojo M, Kuroda T, Ono K, Kimura N, Matsubara E, Osakada Y, Wakutani Y, Takao Y, Higashi Y, Asada K, Senga T, Lee LJ, Tanaka K. A New Serum Biomarker Set to Detect Mild Cognitive Impairment and Alzheimer's Disease by Peptidome Technology. J Alzheimers Dis 2021; 73:217-227. [PMID: 31771070 PMCID: PMC7029318 DOI: 10.3233/jad-191016] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: Because dementia is an emerging problem in the world, biochemical markers of cerebrospinal fluid (CSF) and radio-isotopic analyses are helpful for diagnosing Alzheimer’s disease (AD). Although blood sample is more feasible and plausible than CSF or radiological biomarkers for screening potential AD, measurements of serum amyloid- β (Aβ), plasma tau, and serum antibodies for Aβ1 - 42 are not yet well established. Objective: We aimed to identify a new serum biomarker to detect mild cognitive impairment (MCI) and AD in comparison to cognitively healthy control by a new peptidome technology. Methods: With only 1.5μl of serum, we examined a new target plate “BLOTCHIP®” plus a matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF/MS) to discriminate control (n = 100), MCI (n = 60), and AD (n = 99). In some subjects, cognitive Mini-Mental State Examination (MMSE) were compared to positron emission tomography (PET) with Pittsburgh compound B (PiB) and the serum probability of dementia (SPD). The mother proteins of candidate serum peptides were examined in autopsied AD brains. Results: Apart from Aβ or tau, the present study discovered a new diagnostic 4-peptides-set biomarker for discriminating control, MCI, and AD with 87% of sensitivity and 65% of specificity between control and AD (***p < 0.001). MMSE score was well correlated to brain Aβ deposition and to SPD of AD. The mother proteins of the four peptides were upregulated for coagulation, complement, and plasticity (three proteins), and was downregulated for anti-inflammation (one protein) in AD brains. Conclusion: The present serum biomarker set provides a new, rapid, non-invasive, highly quantitative and low-cost clinical application for dementia screening, and also suggests an alternative pathomechanism of AD for neuroinflammation and neurovascular unit damage.
Collapse
Affiliation(s)
- Koji Abe
- Department of Neurology, Okayama University, Okayama, Japan
| | - Jingwei Shang
- Department of Neurology, Okayama University, Okayama, Japan
| | - Xiaowen Shi
- Department of Neurology, Okayama University, Okayama, Japan
| | - Toru Yamashita
- Department of Neurology, Okayama University, Okayama, Japan
| | | | - Mami Takemoto
- Department of Neurology, Okayama University, Okayama, Japan
| | - Ryuta Morihara
- Department of Neurology, Okayama University, Okayama, Japan
| | - Yumiko Nakano
- Department of Neurology, Okayama University, Okayama, Japan
| | - Yasuyuki Ohta
- Department of Neurology, Okayama University, Okayama, Japan
| | - Kentaro Deguchi
- Department of Neurology, Okayama City Hospital, Okayama, Japan
| | - Masaki Ikeda
- Department of Neurology, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Yoshio Ikeda
- Department of Neurology, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Koichi Okamoto
- Department of Neurology, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Mikio Shoji
- Department of Neurology, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Masamitsu Takatama
- Department of Neurology, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Motohisa Kojo
- Department of Neurology, Ako Chuo Hospital, Ako, Japan
| | - Takeshi Kuroda
- Division of Neurology, Department of Medicine, Showa University, School of Medicine, Tokyo, Japan
| | - Kenjiro Ono
- Division of Neurology, Department of Medicine, Showa University, School of Medicine, Tokyo, Japan
| | - Noriyuki Kimura
- Department of Neurology, Faculty of Medicine, Oita University, Oita, Japan
| | - Etsuro Matsubara
- Department of Neurology, Faculty of Medicine, Oita University, Oita, Japan
| | - Yosuke Osakada
- Department of Neurology, Kurashiki Heisei Hospital, Kurashiki, Japan
| | - Yosuke Wakutani
- Department of Neurology, Kurashiki Heisei Hospital, Kurashiki, Japan
| | - Yoshiki Takao
- Department of Neurology, Kurashiki Heisei Hospital, Kurashiki, Japan
| | - Yasuto Higashi
- Department of Neurology, Himeji Central Hospital, Himeji, Japan
| | - Kyoichi Asada
- Membrane Protein and Ligand Analysis Center, Protosera Inc., Osaka, Japan
| | - Takehito Senga
- Membrane Protein and Ligand Analysis Center, Protosera Inc., Osaka, Japan
| | - Lyang-Ja Lee
- Membrane Protein and Ligand Analysis Center, Protosera Inc., Osaka, Japan
| | - Kenji Tanaka
- Membrane Protein and Ligand Analysis Center, Protosera Inc., Osaka, Japan
| |
Collapse
|
88
|
Leuzy A, Cullen NC, Mattsson-Carlgren N, Hansson O. Current advances in plasma and cerebrospinal fluid biomarkers in Alzheimer's disease. Curr Opin Neurol 2021; 34:266-274. [PMID: 33470669 DOI: 10.1097/wco.0000000000000904] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW This review provides a concise overview of recent advances in cerebrospinal fluid (CSF) and blood-based biomarkers of Alzheimer's disease lesions. RECENT FINDINGS Important recent advances for CSF Alzheimer's disease biomarkers include the introduction of fully automated assays, the development and implementation of certified reference materials for CSF Aβ42 and a unified protocol for handling of samples, which all support reliability and availability of CSF Alzheimer's disease biomarkers. Aβ deposition can be detected using Aβ42/Aβ40 ratio in both CSF and plasma, though a much more modest change is seen in plasma. Tau aggregation can be detected using phosphorylated tau (P-tau) at threonine 181 and 217 in CSF, with similar accuracy in plasma. Neurofilament light (NfL) be measured in CSF and shows similar diagnostic accuracy in plasma. Though total tau (T-tau) can also be measured in plasma, this measure is of limited clinical relevance for Alzheimer's disease in its current immunoassay format. SUMMARY Alzheimer's disease biomarkers, including Aβ, P-tau and NfL can now be reliably measured in both CSF and blood. Plasma-based measures of P-tau show particular promise, with potential applications in both clinical practice and in clinical trials.
Collapse
Affiliation(s)
- Antoine Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö
| | - Nicholas C Cullen
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö
- Department of Neurology, Skåne University Hospital
- Wallenberg Centre for Molecular Medicine, Lund University
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö
- Memory Clinic, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
89
|
Chong JR, Ashton NJ, Karikari TK, Tanaka T, Saridin FN, Reilhac A, Robins EG, Nai YH, Vrooman H, Hilal S, Zetterberg H, Blennow K, Lai MKP, Chen CP. Plasma P-tau181 to Aβ42 ratio is associated with brain amyloid burden and hippocampal atrophy in an Asian cohort of Alzheimer's disease patients with concomitant cerebrovascular disease. Alzheimers Dement 2021; 17:1649-1662. [PMID: 33792168 DOI: 10.1002/alz.12332] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/01/2021] [Accepted: 02/18/2021] [Indexed: 12/20/2022]
Abstract
INTRODUCTION There is increasing evidence that phosphorylated tau (P-tau181) is a specific biomarker for Alzheimer's disease (AD) pathology, but its potential utility in non-White patient cohorts and patients with concomitant cerebrovascular disease (CeVD) is unknown. METHODS Single molecule array (Simoa) measurements of plasma P-tau181, total tau, amyloid beta (Aβ)40 and Aβ42, as well as derived ratios were correlated with neuroimaging modalities indicating brain amyloid (Aβ+), hippocampal atrophy, and CeVD in a Singapore-based cohort of non-cognitively impaired (NCI; n = 43), cognitively impaired no dementia (CIND; n = 91), AD (n = 44), and vascular dementia (VaD; n = 22) subjects. RESULTS P-tau181/Aβ42 ratio showed the highest area under the curve (AUC) for Aβ+ (AUC = 0.889) and for discriminating between AD Aβ+ and VaD Aβ- subjects (AUC = 0.903). In addition, P-tau181/Aβ42 ratio was associated with hippocampal atrophy. None of the biomarkers was associated with CeVD. DISCUSSION Plasma P-tau181/Aβ42 ratio may be a noninvasive means of identifying AD with elevated brain amyloid in populations with concomitant CeVD.
Collapse
Affiliation(s)
- Joyce R Chong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore.,Memory, Aging and Cognition Centre, National University Health Systems, Kent Ridge, Singapore
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Psychology and Neuroscience, King's College London, Institute of Psychiatry, 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
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Tomotaka Tanaka
- Memory, Aging and Cognition Centre, National University Health Systems, Kent Ridge, Singapore.,Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan.,Clinical Imaging Research Centre, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - Francis N Saridin
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore.,Memory, Aging and Cognition Centre, National University Health Systems, Kent Ridge, Singapore
| | - Anthonin Reilhac
- Clinical Imaging Research Centre, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - Edward G Robins
- Clinical Imaging Research Centre, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore.,Technology and Research, Biopolis, Singapore Bioimaging Consortium, A*Star Agency for Science, Singapore
| | - Ying-Hwey Nai
- Clinical Imaging Research Centre, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - Henri Vrooman
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Saima Hilal
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore.,Memory, Aging and Cognition Centre, National University Health Systems, Kent Ridge, Singapore.,Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Kent Ridge, Singapore
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,UK Dementia Research Institute at UCL, London, UK.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - 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
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore.,Memory, Aging and Cognition Centre, National University Health Systems, Kent Ridge, Singapore
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore.,Memory, Aging and Cognition Centre, National University Health Systems, Kent Ridge, Singapore
| |
Collapse
|
90
|
Tissot C, L Benedet A, Therriault J, Pascoal TA, Lussier FZ, Saha-Chaudhuri P, Chamoun M, Savard M, Mathotaarachchi SS, Bezgin G, Wang YT, Fernandez Arias J, Rodriguez JL, Snellman A, Ashton NJ, Karikari TK, Blennow K, Zetterberg H, De Villers-Sidani E, Huot P, Gauthier S, Rosa-Neto P. Plasma pTau181 predicts cortical brain atrophy in aging and Alzheimer's disease. Alzheimers Res Ther 2021; 13:69. [PMID: 33781319 PMCID: PMC8008680 DOI: 10.1186/s13195-021-00802-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 03/08/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND To investigate the association of plasma pTau181, assessed with a new immunoassay, with neurodegeneration of white matter and gray matter cross-sectionally and longitudinally, in aging and Alzheimer's disease. METHODS Observational data was obtained from the Alzheimer's Disease Neuroimaging Initiative, in which participants underwent plasma assessment and magnetic resonance imaging. Based on their clinical diagnosis, participants were classified as cognitively unimpaired and cognitively impaired. Linear regressions and linear mixed-effect models were used to test the cross-sectional and longitudinal associations between baseline plasma pTau181 and neurodegeneration using voxel-based morphometry. RESULTS We observed a negative correlation at baseline between plasma pTau181 and gray matter volume in cognitively unimpaired individuals. In cognitively impaired individuals, we observed a negative association between plasma pTau181 and both gray and white matter volume. In longitudinal analyses conducted in the cognitively unimpaired group, plasma pTau181 was negatively correlated with gray matter volume, starting 36 months after baseline assessments. Finally, in cognitively impaired individuals, plasma pTau181 concentrations were negatively correlated with both gray and white matter volume as early as 12 months after baseline, and neurodegeneration increased in an incremental manner until 48 months. CONCLUSIONS Higher levels of plasma pTau181 correlate with neurodegeneration and predict further brain atrophy in aging and Alzheimer's disease. Plasma pTau181 may be useful in predicting AD-related neurodegeneration, comparable to positron emission tomography or cerebrospinal fluid assessment with high specificity for AD neurodegeneration.
Collapse
Affiliation(s)
- Cécile Tissot
- The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 875 La Salle Blvd - FBC room 3149, Montreal, QC, H4H 1R3, Canada
- Translational Neuroimaging Laboratory-McGill University, Montreal, QC, Canada
- Douglas Hospital Research Centre, Verdun, QC, Canada
| | - Andréa L Benedet
- The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 875 La Salle Blvd - FBC room 3149, Montreal, QC, H4H 1R3, Canada
- Translational Neuroimaging Laboratory-McGill University, Montreal, QC, Canada
| | - Joseph Therriault
- The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 875 La Salle Blvd - FBC room 3149, Montreal, QC, H4H 1R3, Canada
- Translational Neuroimaging Laboratory-McGill University, Montreal, QC, Canada
| | - Tharick A Pascoal
- The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 875 La Salle Blvd - FBC room 3149, Montreal, QC, H4H 1R3, Canada
- Translational Neuroimaging Laboratory-McGill University, Montreal, QC, Canada
| | - Firoza Z Lussier
- The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 875 La Salle Blvd - FBC room 3149, Montreal, QC, H4H 1R3, Canada
- Translational Neuroimaging Laboratory-McGill University, Montreal, QC, Canada
| | | | - Mira Chamoun
- The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 875 La Salle Blvd - FBC room 3149, Montreal, QC, H4H 1R3, Canada
- Translational Neuroimaging Laboratory-McGill University, Montreal, QC, Canada
| | - Melissa Savard
- The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 875 La Salle Blvd - FBC room 3149, Montreal, QC, H4H 1R3, Canada
- Translational Neuroimaging Laboratory-McGill University, Montreal, QC, Canada
| | - Sulantha S Mathotaarachchi
- The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 875 La Salle Blvd - FBC room 3149, Montreal, QC, H4H 1R3, Canada
- Translational Neuroimaging Laboratory-McGill University, Montreal, QC, Canada
| | - Gleb Bezgin
- The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 875 La Salle Blvd - FBC room 3149, Montreal, QC, H4H 1R3, Canada
- Translational Neuroimaging Laboratory-McGill University, Montreal, QC, Canada
| | - Yi-Ting Wang
- The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 875 La Salle Blvd - FBC room 3149, Montreal, QC, H4H 1R3, Canada
- Translational Neuroimaging Laboratory-McGill University, Montreal, QC, Canada
| | - Jaime Fernandez Arias
- The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 875 La Salle Blvd - FBC room 3149, Montreal, QC, H4H 1R3, Canada
- Translational Neuroimaging Laboratory-McGill University, Montreal, QC, Canada
| | - Juan Lantero Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anniina Snellman
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- 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
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, 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
| | - 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
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | | | - Philippe Huot
- Neurodegenerative disease groups, Montreal Neurological Institute, Montreal, QC, Canada
| | - Serge Gauthier
- The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 875 La Salle Blvd - FBC room 3149, Montreal, QC, H4H 1R3, Canada
- Douglas Hospital Research Centre, Verdun, QC, Canada
| | - Pedro Rosa-Neto
- The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 875 La Salle Blvd - FBC room 3149, Montreal, QC, H4H 1R3, Canada.
- Translational Neuroimaging Laboratory-McGill University, Montreal, QC, Canada.
- Douglas Hospital Research Centre, Verdun, QC, Canada.
- Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, QC, Canada.
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Canada.
| |
Collapse
|
91
|
Clark C, Lewczuk P, Kornhuber J, Richiardi J, Maréchal B, Karikari TK, Blennow K, Zetterberg H, Popp J. Plasma neurofilament light and phosphorylated tau 181 as biomarkers of Alzheimer's disease pathology and clinical disease progression. ALZHEIMERS RESEARCH & THERAPY 2021; 13:65. [PMID: 33766131 PMCID: PMC7995778 DOI: 10.1186/s13195-021-00805-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/09/2021] [Indexed: 01/01/2023]
Abstract
Background To assess the performance of plasma neurofilament light (NfL) and phosphorylated tau 181 (p-tau181) to inform about cerebral Alzheimer’s disease (AD) pathology and predict clinical progression in a memory clinic setting. Methods Plasma NfL and p-tau181, along with established cerebrospinal fluid (CSF) biomarkers of AD pathology, were measured in participants with normal cognition (CN) and memory clinic patients with cognitive impairment (mild cognitive impairment and dementia, CI). Clinical and neuropsychological assessments were performed at inclusion and follow-up visits at 18 and 36 months. Multivariate analysis assessed associations of plasma NfL and p-tau181 levels with AD, single CSF biomarkers, hippocampal volume, and clinical measures of disease progression. Results Plasma NfL levels were higher in CN participants with an AD CSF profile (defined by a CSF p-tau181/Aβ1–42 > 0.0779) as compared with CN non-AD, while p-tau181 plasma levels were higher in CI patients with AD. Plasma NfL levels correlated with CSF tau and p-tau181 in CN, and with CSF tau in CI patients. Plasma p-tau181 correlated with CSF p-tau181 in CN and with CSF tau, p-tau181, Aβ1–42, and Aβ1–42/Aβ1–40 in CI participants. Compared with a reference model, adding plasma p-tau181 improved the prediction of AD in CI patients while adding NfL did not. Adding p-tau181, but not NfL levels, to a reference model improved prediction of cognitive decline in CI participants. Conclusion Plasma NfL indicates neurodegeneration while plasma p-tau181 levels can serve as a biomarker of cerebral AD pathology and cognitive decline. Their predictive performance depends on the presence of cognitive impairment.
Collapse
Affiliation(s)
- Christopher Clark
- Institute for Regenerative Medicine, University of Zürich, Zürich, Switzerland.
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich - Alexander Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Neurodegeneration Diagnostics, Medical University of Białystok, Białystok, Poland
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich - Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jonas Richiardi
- Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Bénédicte Maréchal
- Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.,Advanced Clinical Imaging Technology group, Siemens Healthcare AG, Lausanne, Switzerland.,LTS5, École Polytechnique FÉdÉrale de Lausanne (EPFL), Lausanne, Switzerland
| | - Thomas K Karikari
- 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
| | - 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
| | - 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.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Julius Popp
- Old age Psychiatry, Department of Psychiatry, University Hospital of Lausanne, Lausanne, Switzerland.,Department of Geriatric Psychiatry, University Hospital of Psychiatry Zürich and University of Zürich, Zürich, Switzerland
| |
Collapse
|
92
|
Ding X, Zhang S, Jiang L, Wang L, Li T, Lei P. Ultrasensitive assays for detection of plasma tau and phosphorylated tau 181 in Alzheimer's disease: a systematic review and meta-analysis. Transl Neurodegener 2021; 10:10. [PMID: 33712071 PMCID: PMC7953695 DOI: 10.1186/s40035-021-00234-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/24/2021] [Indexed: 02/08/2023] Open
Abstract
A lack of convenient and reliable biomarkers for diagnosis and prognosis is a common challenge for neurodegenerative diseases such as Alzheimer's disease (AD). Recent advancement in ultrasensitive protein assays has allowed the quantification of tau and phosphorylated tau proteins in peripheral plasma. Here we identified 66 eligible studies reporting quantification of plasma tau and phosphorylated tau 181 (ptau181) using four ultrasensitive methods. Meta-analysis of these studies confirmed that the AD patients had significantly higher plasma tau and ptau181 levels compared with controls, and that the plasma tau and ptau181 could predict AD with high-accuracy area under curve of the Receiver Operating Characteristic. Therefore, plasma tau and plasma ptau181 can be considered as biomarkers for AD diagnosis.
Collapse
Affiliation(s)
- Xulong Ding
- Department of Neurology and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shuting Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lijun Jiang
- Mental Health Center and West China Brain Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lu Wang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tao Li
- Mental Health Center and West China Brain Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
93
|
Barthélemy NR, Horie K, Sato C, Bateman RJ. Blood plasma phosphorylated-tau isoforms track CNS change in Alzheimer's disease. J Exp Med 2021; 217:151982. [PMID: 32725127 PMCID: PMC7596823 DOI: 10.1084/jem.20200861] [Citation(s) in RCA: 251] [Impact Index Per Article: 83.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/15/2020] [Accepted: 06/24/2020] [Indexed: 01/31/2023] Open
Abstract
Highly sensitive and specific plasma biomarkers for Alzheimer’s disease (AD) have the potential to improve diagnostic accuracy in the clinic and facilitate research studies including enrollment in prevention and treatment trials. We recently reported CSF tau hyperphosphorylation, especially on T217, is an accurate predictor of β-amyloidosis at asymptomatic and symptomatic stages. In the current study, we determine by mass spectrometry the potential utility of plasma p-tau isoforms to detect AD pathology and investigate CSF and plasma tau isoforms’ profile relationships. Plasma tau was truncated as previously described in CSF. CSF and plasma measures of p-tau-217 and p-tau-181 were correlated. No correlation was found between CSF and plasma on total-tau levels and pS202 measures. We found p-tau-217 and p-tau-181 were highly specific for amyloid plaque pathology in the discovery cohort (n = 36, AUROC = 0.99 and 0.98 respectively). In the validation cohort (n = 92), p-tau-217 measures were still specific to amyloid status (AUROC = 0.92), and p-tau-181 measures were less specific (AUROC = 0.75).
Collapse
Affiliation(s)
- Nicolas R Barthélemy
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Kanta Horie
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Chihiro Sato
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO.,Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
94
|
Aslan JE. Platelet Proteomes, Pathways, and Phenotypes as Informants of Vascular Wellness and Disease. Arterioscler Thromb Vasc Biol 2021; 41:999-1011. [PMID: 33441027 PMCID: PMC7980774 DOI: 10.1161/atvbaha.120.314647] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Platelets rapidly undergo responsive transitions in form and function to repair vascular endothelium and mediate hemostasis. In contrast, heterogeneous platelet subpopulations with a range of primed or refractory phenotypes gradually arise in chronic inflammatory and other conditions in a manner that may indicate or support disease. Qualitatively distinguishable platelet phenotypes are increasingly associated with a variety of physiological and pathological circumstances; however, the origins and significance of platelet phenotypic variation remain unclear and conceptually vague. As changes in platelet function in disease exhibit many similarities to platelets following the activation of platelet agonist receptors, the intracellular responses of platelets common to hemostasis and inflammation may provide insights to the molecular basis of platelet phenotype. Here, we review concepts around how protein-level relations-from platelet receptors through intracellular signaling events-may help to define platelet phenotypes in inflammation, immune responses, aging, and other conditions. We further discuss how representing systems-wide platelet proteomics data profiles as circuit-like networks of causally related intracellular events, or, pathway maps, may inform molecular definitions of platelet phenotype. In addition to offering insights into platelets as druggable targets, maps of causally arranged intracellular relations underlying platelet function can also advance precision and interceptive medicine efforts by leveraging platelets as accessible, dynamic, endogenous, circulating biomarkers of vascular wellness and disease. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Joseph E. Aslan
- Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Department of Chemical Physiology and Biochemistry and School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
95
|
Zetterberg H, Blennow K. Moving fluid biomarkers for Alzheimer's disease from research tools to routine clinical diagnostics. Mol Neurodegener 2021; 16:10. [PMID: 33608044 PMCID: PMC7893769 DOI: 10.1186/s13024-021-00430-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/05/2021] [Indexed: 12/31/2022] Open
Abstract
Four fluid-based biomarkers have been developed into diagnostic tests for Alzheimer’s disease (AD) pathology: the ratio of 42 to 40 amino acid-long amyloid β, a marker of plaque pathology; total-tau and phosphorylated tau, markers of AD-related changes in tau metabolism and secretion; and neurofilament light, a marker of neurodegeneration. When measured in cerebrospinal fluid, these biomarkers can be used in clinical practice to support a diagnosis of mild cognitive impairment or dementia due to AD. Recently, technological breakthroughs have made it possible to measure them in standard blood samples as well. Here, we give an updated account of the current state of the fluid-based AD biomarker research field. We discuss how the new blood tests may be used in research and clinical practice, and what role they may play in relation to more established diagnostic tests, such as CSF biomarkers and amyloid and tau positron emission tomography, to facilitate the effective implementation of future disease-modifying therapies.
Collapse
Affiliation(s)
- Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy, 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.
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden. .,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
| |
Collapse
|
96
|
Arbaciauskaite M, Lei Y, Cho YK. High-specificity antibodies and detection methods for quantifying phosphorylated tau from clinical samples. Antib Ther 2021; 4:34-44. [PMID: 33928234 PMCID: PMC7944500 DOI: 10.1093/abt/tbab004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/14/2021] [Accepted: 02/05/2021] [Indexed: 12/14/2022] Open
Abstract
The ability to measure total and phosphorylated tau levels in clinical samples is transforming the detection of Alzheimer’s disease (AD) and other neurodegenerative diseases. In particular, recent reports indicate that accurate detection of low levels of phosphorylated tau (p-tau) in plasma provides a reliable biomarker of AD long before sensing memory loss. Therefore, the diagnosis and monitoring of neurodegenerative diseases progression using blood samples is becoming a reality. These major advances were achieved by using antibodies specific to p-tau as well as sophisticated high-sensitivity immunoassay platforms. This review focuses on these enabling advances in high-specificity antibody development, engineering, and novel signal detection methods. We will draw insights from structural studies on p-tau antibodies, engineering efforts to improve their binding properties, and efforts to validate their specificity. A comprehensive survey of high-sensitivity p-tau immunoassay platforms along with sensitivity limits will be provided. We conclude that although robust approaches for detecting certain p-tau species have been established, systematic efforts to validate antibodies for assay development is still needed for the recognition of biomarkers for AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Monika Arbaciauskaite
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Yu Lei
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Yong Ku Cho
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
97
|
Janelidze S, Berron D, Smith R, Strandberg O, Proctor NK, Dage JL, Stomrud E, Palmqvist S, Mattsson-Carlgren N, Hansson O. Associations of Plasma Phospho-Tau217 Levels With Tau Positron Emission Tomography in Early Alzheimer Disease. JAMA Neurol 2021; 78:149-156. [PMID: 33165506 PMCID: PMC7653537 DOI: 10.1001/jamaneurol.2020.4201] [Citation(s) in RCA: 185] [Impact Index Per Article: 61.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Importance There is an urgent need for inexpensive and minimally invasive blood biomarkers for Alzheimer disease (AD) that could be used to detect early disease changes. Objective To assess how early in the course of AD plasma levels of tau phosphorylated at threonine 217 (P-tau217) start to change compared with levels of established cerebrospinal fluid (CSF) and positron emission tomography (PET) biomarkers of AD pathology. Design, Setting, and Participants This cohort study included cognitively healthy control individuals (n = 225) and participants with subjective cognitive decline (n = 89) or mild cognitive impairment (n = 176) from the BioFINDER-2 study. Participants were enrolled at 2 different hospitals in Sweden from January 2017 to October 2019. All study participants underwent plasma P-tau217 assessments and tau- and amyloid-β (Aβ)-PET imaging. A subcohort of 111 participants had 2 or 3 tau-PET scans. Main Outcomes and Measures Changes in plasma P-tau217 levels in preclinical and prodromal AD compared with changes in CSF P-tau217 and PET measures. Results Of 490 participants, 251 were women (51.2%) and the mean (SD) age was 65.9 (13.1) years. Plasma P-tau217 levels were increased in cognitively unimpaired participants with abnormal Aβ-PET but normal tau-PET in the entorhinal cortex (Aβ-PET+/ tau-PET- group vs Aβ-PET-/ tau-PET- group: median, 2.2 pg/mL [interquartile range (IQR), 1.5-2.9 pg/mL] vs 0.7 pg/mL [IQR, 0.3-1.4 pg/mL]). Most cognitively unimpaired participants who were discordant for plasma P-tau217 and tau-PET were positive for plasma P-tau217 and negative for tau-PET (P-tau217+/tau-PET-: 36 [94.7%]; P-tau217-/tau-PET+: 2 [5.3%]). Event-based modeling of cross-sectional data predicted that in cognitively unimpaired participants and in those with mild cognitive impairment, both plasma and CSF P-tau217 would change before the tau-PET signal in the entorhinal cortex, followed by more widespread cortical tau-PET changes. When testing the association with global Aβ load in nonlinear spline models, both plasma and CSF P-tau217 were increased at lower Aβ-PET values compared with tau-PET measures. Among participants with normal baseline tau-PET, the rates of longitudinal increase in tau-PET in the entorhinal cortex were higher in those with abnormal plasma P-tau217 at baseline (median standardized uptake value ratio, 0.029 [IQR, -0.006 to 0.041] vs -0.001 [IQR, -0.021 to 0.020]; Mann-Whitney U, P = .02). Conclusions and Relevance In this cohort study, plasma P-tau217 levels were increased during the early preclinical stages of AD when insoluble tau aggregates were not yet detectable by tau-PET. Plasma P-tau217 may hold promise as a biomarker for early AD brain pathology.
Collapse
Affiliation(s)
- Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sölvegatan, Sweden
| | - David Berron
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sölvegatan, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sölvegatan, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sölvegatan, Sweden
| | | | | | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sölvegatan, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sölvegatan, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sölvegatan, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sölvegatan, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
98
|
Karikari TK, Benedet AL, Ashton NJ, Lantero Rodriguez J, Snellman A, Suárez-Calvet M, Saha-Chaudhuri P, Lussier F, Kvartsberg H, Rial AM, Pascoal TA, Andreasson U, Schöll M, Weiner MW, Rosa-Neto P, Trojanowski JQ, Shaw LM, Blennow K, Zetterberg H. Diagnostic performance and prediction of clinical progression of plasma phospho-tau181 in the Alzheimer's Disease Neuroimaging Initiative. Mol Psychiatry 2021; 26:429-442. [PMID: 33106600 DOI: 10.1038/s41380-020-00923-z] [Citation(s) in RCA: 188] [Impact Index Per Article: 62.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/01/2020] [Accepted: 10/08/2020] [Indexed: 11/10/2022]
Abstract
Whilst cerebrospinal fluid (CSF) and positron emission tomography (PET) biomarkers for amyloid-β (Aβ) and tau pathologies are accurate for the diagnosis of Alzheimer's disease (AD), their broad implementation in clinical and trial settings are restricted by high cost and limited accessibility. Plasma phosphorylated-tau181 (p-tau181) is a promising blood-based biomarker that is specific for AD, correlates with cerebral Aβ and tau pathology, and predicts future cognitive decline. In this study, we report the performance of p-tau181 in >1000 individuals from the Alzheimer's Disease Neuroimaging Initiative (ADNI), including cognitively unimpaired (CU), mild cognitive impairment (MCI) and AD dementia patients characterized by Aβ PET. We confirmed that plasma p-tau181 is increased at the preclinical stage of Alzheimer and further increases in MCI and AD dementia. Individuals clinically classified as AD dementia but having negative Aβ PET scans show little increase but plasma p-tau181 is increased if CSF Aβ has already changed prior to Aβ PET changes. Despite being a multicenter study, plasma p-tau181 demonstrated high diagnostic accuracy to identify AD dementia (AUC = 85.3%; 95% CI, 81.4-89.2%), as well as to distinguish between Aβ- and Aβ+ individuals along the Alzheimer's continuum (AUC = 76.9%; 95% CI, 74.0-79.8%). Higher baseline concentrations of plasma p-tau181 accurately predicted future dementia and performed comparably to the baseline prediction of CSF p-tau181. Longitudinal measurements of plasma p-tau181 revealed low intra-individual variability, which could be of potential benefit in disease-modifying trials seeking a measurable response to a therapeutic target. This study adds significant weight to the growing body of evidence in the use of plasma p-tau181 as a non-invasive diagnostic and prognostic tool for AD, regardless of clinical stage, which would be of great benefit in clinical practice and a large cost-saving in clinical trial recruitment.
Collapse
Affiliation(s)
- Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Andréa L Benedet
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, H4H 1R3, Montreal, QC, Canada
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Mölndal, Sweden.,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
| | - Juan Lantero Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Anniina Snellman
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.,Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Servei de Neurologia, Hospital del Mar, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | | | - Firoza Lussier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, H4H 1R3, Montreal, QC, Canada
| | - Hlin Kvartsberg
- 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
| | - Alexis Moscoso Rial
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Mölndal, Sweden
| | - Tharick A Pascoal
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, H4H 1R3, Montreal, QC, Canada.,Montreal Neurological Institute, H3A 2B4, Montreal, QC, Canada
| | - Ulf Andreasson
- 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
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Michael W Weiner
- Department of Radiology, Medicine, and Psychiatry, University of California San Francisco, San Francisco, CA, USA
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, H4H 1R3, Montreal, QC, Canada.,Montreal Neurological Institute, H3A 2B4, Montreal, QC, Canada.,Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Institute on Aging, Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Leslie M Shaw
- Institute on Aging, Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA, 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
| | - 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 Institute of Neurology, London, UK. .,UK Dementia Research Institute at UCL, London, UK.
| | | |
Collapse
|
99
|
Shi Y, Gu L, Wang Q, Gao L, Zhu J, Lu X, Zhou F, Zhu D, Zhang H, Xie C, Zhang Z. Platelet Amyloid-β Protein Precursor (AβPP) Ratio and Phosphorylated Tau as Promising Indicators for Early Alzheimer's Disease. J Gerontol A Biol Sci Med Sci 2021; 75:664-670. [PMID: 31336382 DOI: 10.1093/gerona/glz005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Indexed: 12/19/2022] Open
Abstract
To identify whether platelet amyloid-β protein precursor (AβPP) ratio, phosphorylated-tau (P-tau) 231, P-tau181, and serine 396 and 404 (Ser396/404) phosphorylated tau are potential peripheral indicators for early Alzheimer's disease (AD). Forty-three amnesic mild cognitive impairment (aMCI) patients and 45 normal controls were recruited. Peripheral venous blood was drawn and platelets were collected and evaluated for potential indicators by Western blot analysis. Subsequent meta-analysis was completed on these selected indicators. In platelets of aMCI patients, the AβPP ratio level was significantly lower and levels of P-tau231 and Ser396/404 phosphorylated tau were significantly higher. Moreover, in aMCI patients, a negative correlation was observed between platelet P-tau231 level and the Trail Making Tests A score, and it was found that higher platelet P-tau231 levels significantly associated with a worse performance of information processing speed. Furthermore, values of the area under the curve of platelet P-tau231 and Ser396/404 phosphorylated tau were 0.624 and 0.657, respectively. Finally, a meta-analysis indicated platelet AβPP ratio level was significantly lower in MCI cohorts. In conclusion, platelets of aMCI subjects showed a lower AβPP ratio and higher levels of P-tau231 and Ser396/404 phosphorylated tau when compared to normal controls, which may be critical in identifying early AD.
Collapse
Affiliation(s)
- Yachen Shi
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Lihua Gu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Qing Wang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Lijuan Gao
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Jianli Zhu
- Department of Psychology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiang Lu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Fangfang Zhou
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Dan Zhu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Haisan Zhang
- Department of Psychology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Chunming Xie
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Zhijun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China.,Department of Psychology, Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
100
|
Petersen ME, Rafii MS, Zhang F, Hall J, Julovich D, Ances BM, Schupf N, Krinsky-McHale SJ, Mapstone M, Silverman W, Lott I, Klunk W, Head E, Christian B, Foroud T, Lai F, Rosas HD, Zaman S, Wang MC, Tycko B, Lee JH, Handen B, Hartley S, Fortea J, O’Bryant S. Plasma Total-Tau and Neurofilament Light Chain as Diagnostic Biomarkers of Alzheimer's Disease Dementia and Mild Cognitive Impairment in Adults with Down Syndrome. J Alzheimers Dis 2021; 79:671-681. [PMID: 33337378 PMCID: PMC8273927 DOI: 10.3233/jad-201167] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The need for diagnostic biomarkers of cognitive decline is particularly important among aging adults with Down syndrome (DS). Growing empirical support has identified the utility of plasma derived biomarkers among neurotypical adults with mild cognitive impairment (MCI) and Alzheimer's disease (AD); however, the application of such biomarkers has been limited among the DS population. OBJECTIVE This study aimed to investigate the cross-sectional diagnostic performance of plasma neurofilament light chain (Nf-L) and total-tau, individually and in combination among a cohort of DS adults. METHODS Plasma samples were analyzed from n = 305 (n = 225 cognitively stable (CS); n = 44 MCI-DS; n = 36 DS-AD) participants enrolled in the Alzheimer's Biomarker Consortium -Down Syndrome. RESULTS In distinguishing DS-AD participants from CS, Nf-L alone produced an AUC of 90%, total-tau alone reached 74%, and combined reached an AUC of 86%. When age and gender were included, AUC increased to 93%. Higher values of Nf-L, total-tau, and age were all shown to be associated with increased risk for DS-AD. When distinguishing MCI-DS participants from CS, Nf-L alone produced an AUC of 65%, while total-tau alone reached 56%. A combined model with Nf-L, total-tau, age, and gender produced an AUC of 87%. Both higher values in age and total-tau were found to increase risk for MCI-DS; Nf-L levels were not associated with increased risk for MCI-DS. CONCLUSION Advanced assay techniques make total-tau and particularly Nf-L useful biomarkers of both AD pathology and clinical status in DS and have the potential to serve as outcome measures in clinical trials for future disease-modifying drugs.
Collapse
Affiliation(s)
- Melissa E. Petersen
- University of North Texas Health Science Center, Department of Family Medicine and Institute for Translational Research, 3500 Camp Bowie Blvd, Fort Worth, Texas, 76107, USA
| | - Michael S. Rafii
- Alzheimer’s Therapeutic Research Institute (ATRI), Keck School of Medicine, University of Southern California, 9860 Mesa Rim Road, San Diego, California, 92121, USA
| | - Fan Zhang
- University of North Texas Health Science Center, Department of Family Medicine and Institute for Translational Research, 3500 Camp Bowie Blvd, Fort Worth, Texas, 76107, USA
| | - James Hall
- University of North Texas Health Science Center, Institute for Translational Research and Department of Pharmacology and Neuroscience, 3500 Camp Bowie Blvd, Fort Worth, Texas, 76107, USA
| | - David Julovich
- University of North Texas Health Science Center, Institute for Translational Research and Department of Pharmacology and Neuroscience, 3500 Camp Bowie Blvd, Fort Worth, Texas, 76107, USA
| | - Beau M. Ances
- Washington University School of Medicine in St. Louis, Center for Advanced Medicine Neuroscience, 4921 Parkview Place, St. Louis, Missouri, 63110, USA
| | - Nicole Schupf
- Columbia University Irving Medical Center, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain/G.H. Sergievsky Center, 630 W 168th St, New York, New York, 10032, USA
- Columbia University, Mailman School of Public Health, Department of Epidemiology, 722 West 168th Street, New York, New York, 10032, USA
- Columbia University Irving Medical Center, Department of Neurology, Neurological Institute 710 West 168 Street, New York, New York, 10032, USA
- Columbia University Medical Center, Department of Psychiatry, 1051 Riverside Drive, New York, New York, 10032, USA
| | - Sharon J. Krinsky-McHale
- NYS Institute for Basic Research in Developmental Disabilities, Department of Psychology, 1050 Forest Hill Road, Staten Island, New York, 10314, USA
| | - Mark Mapstone
- University of California, Irvine, Department of Neurology, 839 Health Sciences Road, Irvine, California, 92697, USA
| | - Wayne Silverman
- University of California, Irvine, School of Medicine, Department of Pediatrics, 101 The City Drive, Mail Code:4482, Orange, California, 92668, USA
| | - Ira Lott
- University of California, Irvine, School of Medicine, Department of Pediatrics, 101 The City Drive, Mail Code:4482, Orange, California, 92668, USA
| | - William Klunk
- University of Pittsburgh, Department of Psychiatry, 3811 O’Hara St., Pittsburgh, Pennsylvania, 15213, USA
| | - Elizabeth Head
- University of California, Irvine, Department of Pathology, 1261 Gillespie Neuroscience Facility, Irvine, California, 92697, USA
| | - Brad Christian
- University of Wisconsin Madison, Department of Medical Physics and Psychiatry, 1500 Highland Ave, Madison, Wisconsin, 53705, USA
| | - Tatiana Foroud
- Indiana University School of Medicine, Department of Medical & Molecular Genetics, 410 W. 10 Street, Indianapolis, IN. 46202. USA
| | - Florence Lai
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, 149 13 Street, Room 10128, Charlestown, Massachusetts, 02129, USA
| | - H. Diana Rosas
- Massachusetts General Hospital, Departments of Neurology and Radiology, Harvard Medical School, 149 13 Street Room 10126, Charlestown, Massachusetts, 02129, USA
| | - Shahid Zaman
- University of Cambridge, School of Clinical Medicine, Department of Psychiatry, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK
- Cambridgeshire and Peterborough NHS Foundation Trust, Fulbourn Hospital, Cambridge, CB21 5EF, UK
| | - Mei-Cheng Wang
- Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205
| | - Benjamin Tycko
- Columbia University Irving Medical Center, Department of Pathology and Cell Biology, 630 West 168 Street, New York, NY 10032
| | - Joseph H. Lee
- Columbia University Irving Medical Center, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain/G.H. Sergievsky Center, 630 W 168th St, New York, New York, 10032, USA
| | - Benjamin Handen
- University of Pittsburgh, Department of Psychiatry, 3811 O’Hara St., Pittsburgh, Pennsylvania, 15213, USA
| | - Sigan Hartley
- University of Wisconsin, School of Human Ecology and Waisman Center, 1500 Highland Ave, Madison, WI 53705
| | - Juan Fortea
- Barcelona Down Medical Center. Fundació Catalana de Síndrome de Down. Barcelona, Spain
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sid O’Bryant
- University of North Texas Health Science Center, Institute for Translational Research and Department of Pharmacology and Neuroscience, 3500 Camp Bowie Blvd, Fort Worth, Texas, 76107, USA
| |
Collapse
|