7
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Plasma GFAP is an early marker of amyloid-β but not tau pathology in Alzheimer’s disease

      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

          Although recent clinical trials targeting amyloid-β in Alzheimer’s disease have shown promising results, there is increasing evidence suggesting that understanding alternative disease pathways that interact with amyloid-β metabolism and amyloid pathology might be important to halt the clinical deterioration. In particular, there is evidence supporting a critical role of astroglial activation and astrocytosis in Alzheimer’s disease. However, so far, no studies have assessed whether astrocytosis is independently related to either amyloid-β or tau pathology in vivo.

          To address this question, we determined the levels of the astrocytic marker GFAP in plasma and CSF of 217 amyloid-β-negative cognitively unimpaired individuals, 71 amyloid-β-positive cognitively unimpaired individuals, 78 amyloid-β-positive cognitively impaired individuals, 63 amyloid-β-negative cognitively impaired individuals and 75 patients with a non-Alzheimer’s disease neurodegenerative disorder from the Swedish BioFINDER-2 study. Participants underwent longitudinal amyloid-β ( 18F-flutemetamol) and tau ( 18F-RO948) PET as well as cognitive testing.

          We found that plasma GFAP concentration was significantly increased in all amyloid-β-positive groups compared with participants without amyloid-β pathology ( P < 0.01). In addition, there were significant associations between plasma GFAP with higher amyloid-β-PET signal in all amyloid-β-positive groups, but also in cognitively normal individuals with normal amyloid-β values ( P < 0.001), which remained significant after controlling for tau-PET signal. Furthermore, plasma GFAP could predict amyloid-β-PET positivity with an area under the curve of 0.76, which was greater than the performance achieved by CSF GFAP (0.69) and other glial markers (CSF YKL-40: 0.64, soluble TREM2: 0.71). Although correlations were also observed between tau-PET and plasma GFAP, these were no longer significant after controlling for amyloid-β-PET. In contrast to plasma GFAP, CSF GFAP concentration was significantly increased in non-Alzheimer’s disease patients compared to other groups ( P < 0.05) and correlated with amyloid-β-PET only in amyloid-β-positive cognitively impaired individuals ( P = 0.005). Finally, plasma GFAP was associated with both longitudinal amyloid-β-PET and cognitive decline, and mediated the effect of amyloid-β-PET on tau-PET burden, suggesting that astrocytosis secondary to amyloid-β aggregation might promote tau accumulation.

          Altogether, these findings indicate that plasma GFAP is an early marker associated with brain amyloid-β pathology but not tau aggregation, even in cognitively normal individuals with a normal amyloid-β status. This suggests that plasma GFAP should be incorporated in current hypothetical models of Alzheimer’s disease pathogenesis and be used as a non-invasive and accessible tool to detect early astrocytosis secondary to amyloid-β pathology.

          Abstract

          Pereira et al. show that plasma GFAP is an early and independent marker of astrocytosis associated with Aβ pathology. Plasma GFAP correlated with cognitive decline and mediated the effect of Aβ-PET on tau-PET burden, suggesting that astrocytosis secondary to Aβ aggregation may promote tau accumulation.

          Related collections

          Most cited references55

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

          Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing

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

            Neuroinflammation in Alzheimer's disease.

            Increasing evidence suggests that Alzheimer's disease pathogenesis is not restricted to the neuronal compartment, but includes strong interactions with immunological mechanisms in the brain. Misfolded and aggregated proteins bind to pattern recognition receptors on microglia and astroglia, and trigger an innate immune response characterised by release of inflammatory mediators, which contribute to disease progression and severity. Genome-wide analysis suggests that several genes that increase the risk for sporadic Alzheimer's disease encode factors that regulate glial clearance of misfolded proteins and the inflammatory reaction. External factors, including systemic inflammation and obesity, are likely to interfere with immunological processes of the brain and further promote disease progression. Modulation of risk factors and targeting of these immune mechanisms could lead to future therapeutic or preventive strategies for Alzheimer's disease. Copyright © 2015 Elsevier Ltd. All rights reserved.
              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
                Brain
                Brain
                brainj
                Brain
                Oxford University Press
                0006-8950
                1460-2156
                November 2021
                02 July 2021
                02 July 2021
                : 144
                : 11
                : 3505-3516
                Affiliations
                [1 ] Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute , Stockholm, Sweden
                [2 ] Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University , Sweden
                [3 ] Department of Neurology, Skåne University Hospital, Lund University , Lund, Sweden
                [4 ] Wallenberg Center for Molecular Medicine, Lund University , Lund, Sweden
                [5 ] Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers , Amsterdam, The Netherlands
                [6 ] Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg , Mölndal, Sweden
                [7 ] Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital , Mölndal, Sweden
                [8 ] Department of Neurodegenerative Disease, UCL Institute of Neurology , London, UK
                [9 ] UK Dementia Research Institute at UCL , London, UK
                [10 ] Memory Clinic, Skåne University Hospital , Malmö, Sweden
                [11 ] Wallenberg Centre for Molecular and Translational Medicine, The Sahlgrenska Academy at the University of Gothenburg , Gothenburg, Sweden
                [12 ] Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute , London, UK
                [13 ] NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation , London, UK
                Author notes
                Correspondence to: Joana B. Pereira Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society Karolinska Institute, 141 83 Huddinge, Sweden E-mail: joana.pereira@ 123456ki.se
                Correspondence may also be addressed to: Oskar Hansson Clinical Memory Research Unit, Department of Clinical Sciences, Lund University SE-20502 Malmö, Sweden E-mail: Oskar.Hansson@ 123456med.lu.se
                Author information
                https://orcid.org/0000-0002-3579-8804
                https://orcid.org/0000-0001-7147-0112
                https://orcid.org/0000-0002-8885-7724
                https://orcid.org/0000-0002-4061-0837
                https://orcid.org/0000-0003-3930-4354
                Article
                awab223
                10.1093/brain/awab223
                8677538
                34259835
                b60b21a5-fbdf-4b90-b0eb-9388658d7fe1
                © The Author(s) (2021). Published by Oxford University Press on behalf of the Guarantors of Brain.

                This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License ( https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com

                History
                : 08 April 2021
                : 24 May 2021
                : 05 June 2021
                : 13 December 2021
                Page count
                Pages: 12
                Funding
                Funded by: Swedish Research Council, DOI 10.13039/501100004359;
                Award ID: 2016–00906
                Award ID: 2018–02052
                Funded by: Knut and Alice Wallenberg foundation, DOI 10.13039/501100004063;
                Award ID: 2017–0383
                Funded by: Marianne and Marcus Wallenberg foundation, DOI 10.13039/501100011898;
                Award ID: 2015.0125
                Funded by: Strategic Research Area MultiPark;
                Funded by: Swedish Alzheimer Foundation;
                Award ID: AF-939932
                Funded by: Swedish Brain Foundation;
                Award ID: FO2019-0326
                Funded by: The Parkinson foundation of Sweden;
                Award ID: 1280/20
                Funded by: Skåne University Hospital Foundation;
                Award ID: 2020-O000028
                Funded by: Regionalt Forskningsstöd;
                Award ID: 2020–0314
                Funded by: Swedish federal government;
                Award ID: 2018-Projekt0279
                Funded by: Swedish Research Council, DOI 10.13039/501100004359;
                Award ID: #2018–02201
                Funded by: The Center for Medical Innovation;
                Award ID: #20200695
                Funded by: Karolinska Institute, DOI 10.13039/501100004047;
                Funded by: Swedish Research Council, DOI 10.13039/501100004359;
                Award ID: #2017–00915
                Funded by: Alzheimer Drug Discovery Foundation;
                Award ID: #RDAPB-201809–2016615
                Funded by: Swedish Alzheimer Foundation;
                Award ID: #AF-742881
                Funded by: Hjärnfonden, Sweden;
                Award ID: #FO2017-0243
                Funded by: ALF, DOI 10.13039/100001424;
                Award ID: #ALFGBG-715986
                Funded by: European Union Joint Program for Neurodegenerative Disorders;
                Award ID: JPND2019-466–236
                Funded by: Swedish Research Council, DOI 10.13039/501100004359;
                Award ID: #2018–02532
                Funded by: European Research Council, DOI 10.13039/100010663;
                Award ID: #681712
                Funded by: Swedish State Support for Clinical Research;
                Award ID: #ALFGBG-720931
                Funded by: UK Dementia Research Institute, DOI 10.13039/501100017510;
                Categories
                Original Articles
                AcademicSubjects/MED00310
                AcademicSubjects/SCI01870

                Neurosciences
                astrocytosis,gfap,aβ-pet,tau-pet,cognition
                Neurosciences
                astrocytosis, gfap, aβ-pet, tau-pet, cognition

                Comments

                Comment on this article