14
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      High Soluble Amyloid-β 42 Predicts Normal Cognition in Amyloid-Positive Individuals with Alzheimer’s Disease-Causing Mutations

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background:

          In amyloid-positive individuals at risk for Alzheimer’s disease (AD), high soluble 42-amino acid amyloid-β (Aβ 42) levels are associated with normal cognition. It is unknown if this relationship applies longitudinally in a genetic cohort.

          Objective:

          To test the hypothesis that high Aβ 42 preserves normal cognition in amyloid-positive individuals with Alzheimer’s disease (AD)-causing mutations ( APP, PSEN1, or PSEN2) to a greater extent than lower levels of brain amyloid, cerebrospinal fluid (CSF) phosphorylated tau (p-tau), or total tau (t-tau).

          Methods:

          Cognitive progression was defined as any increase in Clinical Dementia Rating (CDR = 0, normal cognition; 0.5, very mild dementia; 1, mild dementia) over 3 years. Amyloid-positivity was defined as a standard uptake value ratio (SUVR) ≥1.42 by Pittsburgh compound-B positron emission tomography (PiB-PET). We used modified Poisson regression models to estimate relative risk (RR), adjusted for age at onset, sex, education, APOE4 status, and duration of follow-up. The results were confirmed with multiple sensitivity analyses, including Cox regression.

          Results:

          Of 232 mutation carriers, 108 were PiB-PET-positive at baseline, with 43 (39.8%) meeting criteria for progression after 3.3±2.0 years. Soluble Aβ 42 levels were higher among CDR non-progressors than CDR progressors. Higher Aβ 42 predicted a lower risk of progression (adjusted RR, 0.36; 95% confidence interval [CI], 0.19–0.67; p = 0.002) better than lower SUVR (RR, 0.81; 95% CI, 0.68–0.96; p = 0.018). CSF Aβ 42 levels predicting lower risk of progression increased with higher SUVR levels.

          Conclusion:

          High CSF Aβ 42 levels predict normal cognition in amyloid-positive individuals with AD-causing genetic mutations.

          Related collections

          Most cited references40

          • Record: found
          • Abstract: not found
          • Article: not found

          The Clinical Dementia Rating (CDR): current version and scoring rules.

          J Morris (1993)
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            The amyloid hypothesis of Alzheimer's disease at 25 years

            Abstract Despite continuing debate about the amyloid β‐protein (or Aβ hypothesis, new lines of evidence from laboratories and clinics worldwide support the concept that an imbalance between production and clearance of Aβ42 and related Aβ peptides is a very early, often initiating factor in Alzheimer's disease (AD). Confirmation that presenilin is the catalytic site of γ‐secretase has provided a linchpin: all dominant mutations causing early‐onset AD occur either in the substrate (amyloid precursor protein, APP) or the protease (presenilin) of the reaction that generates Aβ. Duplication of the wild‐type APP gene in Down's syndrome leads to Aβ deposits in the teens, followed by microgliosis, astrocytosis, and neurofibrillary tangles typical of AD. Apolipoprotein E4, which predisposes to AD in > 40% of cases, has been found to impair Aβ clearance from the brain. Soluble oligomers of Aβ42 isolated from AD patients' brains can decrease synapse number, inhibit long‐term potentiation, and enhance long‐term synaptic depression in rodent hippocampus, and injecting them into healthy rats impairs memory. The human oligomers also induce hyperphosphorylation of tau at AD‐relevant epitopes and cause neuritic dystrophy in cultured neurons. Crossing human APP with human tau transgenic mice enhances tau‐positive neurotoxicity. In humans, new studies show that low cerebrospinal fluid (CSF) Aβ42 and amyloid‐PET positivity precede other AD manifestations by many years. Most importantly, recent trials of three different Aβ antibodies (solanezumab, crenezumab, and aducanumab) have suggested a slowing of cognitive decline in post hoc analyses of mild AD subjects. Although many factors contribute to AD pathogenesis, Aβ dyshomeostasis has emerged as the most extensively validated and compelling therapeutic target.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Tracking pathophysiological processes in Alzheimer's disease: an updated hypothetical model of dynamic biomarkers.

              In 2010, we put forward a hypothetical model of the major biomarkers of Alzheimer's disease (AD). The model was received with interest because we described the temporal evolution of AD biomarkers in relation to each other and to the onset and progression of clinical symptoms. Since then, evidence has accumulated that supports the major assumptions of this model. Evidence has also appeared that challenges some of our assumptions, which has allowed us to modify our original model. Refinements to our model include indexing of individuals by time rather than clinical symptom severity; incorporation of interindividual variability in cognitive impairment associated with progression of AD pathophysiology; modifications of the specific temporal ordering of some biomarkers; and recognition that the two major proteinopathies underlying AD biomarker changes, amyloid β (Aβ) and tau, might be initiated independently in sporadic AD, in which we hypothesise that an incident Aβ pathophysiology can accelerate antecedent limbic and brainstem tauopathy. Copyright © 2013 Elsevier Ltd. All rights reserved.
                Bookmark

                Author and article information

                Journal
                J Alzheimers Dis
                J Alzheimers Dis
                JAD
                Journal of Alzheimer's Disease
                IOS Press (Nieuwe Hemweg 6B, 1013 BG Amsterdam, The Netherlands )
                1387-2877
                1875-8908
                04 October 2022
                25 October 2022
                2022
                : 90
                : 1
                : 333-348
                Affiliations
                [a ]Department of Neurology, James J. and Joan A. Gardner Family Center for Parkinson’s Disease and Movement Disorders, University of Cincinnati , Cincinnati, OH, USA
                [b ]Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Institutet , Stockholm, Sweden
                [c ]Department of Molecular and Translational Medicine, Division of Biostatistics & Epidemiology, Texas Tech University Health Sciences Center , El Paso, TX, USA
                [d ]A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland , Kuopio, Finland
                [e ]Department of Paediatrics, University of Oxford , Oxford, UK
                [f ]MDUK Oxford Neuromuscular Centre, University of Oxford , Oxford, UK
                [g ]Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta , Parkinson and Movement Disorders Unit, Milan, Italy
                [h ]University of Southern California Keck School of Medicine , Los Angeles, CA, USA
                [i ]Department of Neurology, Mayo Clinic , Jacksonville, FL, USA
                [j ]Department of Clinical Neuroscience, Medical School, Osaka City University , Sutoku University, Abenoku, Osaka, Nagaoka, Japan
                [k ]German Center for Neurodegenerative Diseases , Site Munich, Germany
                [l ]Department of Neurology, Ludwig-Maximilians University Munich , Germany
                [m ]Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet , Stockholm, Sweden
                Author notes
                [ 1 ]

                These authors contributed equally to this work.

                [ 2 ]

                These contributed equally to this work and share senior co-authorship.

                [ # ]

                DIAN Consortia Authors listed below the article.

                [* ]Correspondence to: Dr. Andrea Sturchio, MD, Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Institute, 171 76 Stockholm, Sweden. E-mail: andrea.sturchio@ 123456gmail.com .; andrea.sturchio@ 123456ki.se . and Dr. Alberto J. Espay, MD, MSc, Gardner Family Center for Parkinson’s Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, USA. Tel.: +1 513 558 4035; E-mail alberto.espay@ 123456uc.edu .
                Article
                JAD220808
                10.3233/JAD-220808
                9661329
                36120786
                6ed31893-7663-453f-9719-073980cb304d
                © 2022 – The authors. Published by IOS Press

                This is an open access article distributed under the terms of the Creative Commons Attribution Non-Commercial (CC BY-NC 4.0) License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

                History
                : 15 August 2022
                Categories
                Research Article

                alzheimer’s disease,amyloid-β ,atrophy,cognition,fdg-pet
                alzheimer’s disease, amyloid-β , atrophy, cognition, fdg-pet

                Comments

                Comment on this article