Your browser doesn't support javascript.
loading
Association of Phosphorylated Tau Biomarkers With Amyloid Positron Emission Tomography vs Tau Positron Emission Tomography.
Therriault, Joseph; Vermeiren, Marie; Servaes, Stijn; Tissot, Cécile; Ashton, Nicholas J; Benedet, Andréa Lessa; Karikari, Thomas K; Lantero-Rodriguez, Juan; Brum, Wagner S; Lussier, Firoza Z; Bezgin, Gleb; Stevenson, Jenna; Rahmouni, Nesrine; Kunach, Peter; Wang, Yi-Ting; Fernandez-Arias, Jaime; Socualaya, Kely Quispialaya; Macedo, Arthur C; Ferrari-Souza, João Pedro; Ferreira, Pâmela C L; Bellaver, Bruna; Leffa, Douglas T; Zimmer, Eduardo R; Vitali, Paolo; Soucy, Jean-Paul; Triana-Baltzer, Gallen; Kolb, Hartmuth C; Pascoal, Tharick A; Saha-Chaudhuri, Paramita; Gauthier, Serge; Zetterberg, Henrik; Blennow, Kaj; Rosa-Neto, Pedro.
Afiliação
  • Therriault J; Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada.
  • Vermeiren M; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
  • Servaes S; Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada.
  • Tissot C; Erasmus Medical Center, Erasmus University Rotterdam, Rotterdam, the Netherlands.
  • Ashton NJ; Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada.
  • Benedet AL; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
  • Karikari TK; Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada.
  • Lantero-Rodriguez J; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.
  • Brum WS; Wallenberg Centre for Molecular Medicine, University of Gothenburg, Gothenburg, Sweden.
  • Lussier FZ; King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, United Kingdom.
  • Bezgin G; NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, United Kingdom.
  • Stevenson J; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.
  • Rahmouni N; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.
  • Kunach P; Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
  • Wang YT; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.
  • Fernandez-Arias J; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.
  • Socualaya KQ; Department of Pharmacology, Graduate Program in Biological Sciences: Biochemistry and Pharmacology and Therapeutics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
  • Macedo AC; Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada.
  • Ferrari-Souza JP; Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
  • Ferreira PCL; Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada.
  • Bellaver B; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
  • Leffa DT; Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada.
  • Zimmer ER; Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada.
  • Vitali P; Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada.
  • Soucy JP; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
  • Triana-Baltzer G; Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada.
  • Kolb HC; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
  • Pascoal TA; Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada.
  • Saha-Chaudhuri P; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
  • Gauthier S; Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada.
  • Zetterberg H; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
  • Blennow K; Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada.
  • Rosa-Neto P; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
JAMA Neurol ; 80(2): 188-199, 2023 02 01.
Article em En | MEDLINE | ID: mdl-36508198
Importance: The recent proliferation of phosphorylated tau (p-tau) biomarkers has raised questions about their preferential association with the hallmark pathologies of Alzheimer disease (AD): amyloid-ß plaques and tau neurofibrillary tangles. Objective: To determine whether cerebrospinal fluid (CSF) and plasma p-tau biomarkers preferentially reflect cerebral ß-amyloidosis or neurofibrillary tangle aggregation measured with positron emission tomography (PET). Design, Setting, and Participants: This was a cross-sectional study of 2 observational cohorts: the Translational Biomarkers in Aging and Dementia (TRIAD) study, with data collected between October 2017 and August 2021, and the Alzheimer's Disease Neuroimaging Initiative (ADNI), with data collected between September 2015 and November 2019. TRIAD was a single-center study, and ADNI was a multicenter study. Two independent subsamples were derived from TRIAD. The first TRIAD subsample comprised individuals assessed with CSF p-tau (p-tau181, p-tau217, p-tau231, p-tau235), [18F]AZD4694 amyloid PET, and [18F]MK6240 tau PET. The second TRIAD subsample included individuals assessed with plasma p-tau (p-tau181, p-tau217, p-tau231), [18F]AZD4694 amyloid PET, and [18F]MK6240 tau PET. An independent cohort from ADNI comprised individuals assessed with CSF p-tau181, [18F]florbetapir PET, and [18F]flortaucipir PET. Participants were included based on the availability of p-tau and PET biomarker assessments collected within 9 months of each other. Exclusion criteria were a history of head trauma or magnetic resonance imaging/PET safety contraindications. No participants who met eligibility criteria were excluded. Exposures: Amyloid PET, tau PET, and CSF and plasma assessments of p-tau measured with single molecule array (Simoa) assay or enzyme-linked immunosorbent assay. Main Outcomes and Measures: Associations between p-tau biomarkers with amyloid PET and tau PET. Results: A total of 609 participants (mean [SD] age, 66.9 [13.6] years; 347 female [57%]; 262 male [43%]) were included in the study. For all 4 phosphorylation sites assessed in CSF, p-tau was significantly more closely associated with amyloid-PET values than tau-PET values (p-tau181 difference, 13%; 95% CI, 3%-22%; P = .006; p-tau217 difference, 11%; 95% CI, 3%-20%; P = .003; p-tau231 difference, 15%; 95% CI, 5%-22%; P < .001; p-tau235 difference, 9%; 95% CI, 1%-19%; P = .02) . These results were replicated with plasma p-tau181 (difference, 11%; 95% CI, 1%-22%; P = .02), p-tau217 (difference, 9%; 95% CI, 1%-19%; P = .02), p-tau231 (difference, 13%; 95% CI, 3%-24%; P = .009), and CSF p-tau181 (difference, 9%; 95% CI, 1%-21%; P = .02) in independent cohorts. Conclusions and Relevance: Results of this cross-sectional study of 2 observational cohorts suggest that the p-tau abnormality as an early event in AD pathogenesis was associated with amyloid-ß accumulation and highlights the need for careful interpretation of p-tau biomarkers in the context of the amyloid/tau/neurodegeneration, or A/T/(N), framework.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Doença de Alzheimer / Disfunção Cognitiva Tipo de estudo: Clinical_trials / Observational_studies / Risk_factors_studies Limite: Aged / Female / Humans / Male Idioma: En Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Doença de Alzheimer / Disfunção Cognitiva Tipo de estudo: Clinical_trials / Observational_studies / Risk_factors_studies Limite: Aged / Female / Humans / Male Idioma: En Ano de publicação: 2023 Tipo de documento: Article