scholarly journals Callosal circularity as an early marker for Alzheimer's disease

2018 ◽  
Vol 19 ◽  
pp. 516-526 ◽  
Author(s):  
Jeroen Van Schependom ◽  
Ellis Niemantsverdriet ◽  
Dirk Smeets ◽  
Sebastiaan Engelborghs
2019 ◽  
Vol 15 ◽  
pp. P1548-P1549
Author(s):  
Lene Pålhaugen ◽  
Per Selnes ◽  
Carole H. Sudre ◽  
Sandra Tecelao ◽  
Atle Bjornerud ◽  
...  

2006 ◽  
Vol 14 (7S_Part_13) ◽  
pp. P733-P734
Author(s):  
Eleanor Drummond ◽  
Geoffrey Pires ◽  
Frances Prelli ◽  
Sacha McElligott ◽  
Shiron Drusinsky ◽  
...  

2009 ◽  
Vol 29 (5) ◽  
pp. 1008-1012 ◽  
Author(s):  
Mohammad Haris ◽  
Erin McArdle ◽  
Matthew Fenty ◽  
Anup Singh ◽  
Christos Davatzikos ◽  
...  

1992 ◽  
Vol 94 (1) ◽  
pp. 82
Author(s):  
P. Scheltens ◽  
D. Leys ◽  
F. Barkhof ◽  
H.C. Weinstein ◽  
E.C. Wolters ◽  
...  

2018 ◽  
Vol 64 (4) ◽  
pp. 1359-1371 ◽  
Author(s):  
Christian Sandøe Musaeus ◽  
Knut Engedal ◽  
Peter Høgh ◽  
Vesna Jelic ◽  
Morten Mørup ◽  
...  

2014 ◽  
Vol 41 (3) ◽  
pp. 855-865 ◽  
Author(s):  
Yao-Hsiang Shih ◽  
Kuen-Jer Tsai ◽  
Chu-Wan Lee ◽  
Shu-Chu Shiesh ◽  
Wei-Ting Chen ◽  
...  

2005 ◽  
Vol 1 ◽  
pp. S13-S13
Author(s):  
Paolo Prolo ◽  
Paola Perotti ◽  
Marisa Pautasso ◽  
Maria Luisa Sartori ◽  
Thomas Faccalini ◽  
...  

2020 ◽  
Author(s):  
Elena Tsolaki ◽  
Lajos Csincsik ◽  
Jing Xue ◽  
Imre Lengyel ◽  
Sergio Bertazzo

ABSTRACTDeposition of tau-tangles is an early marker for the development of Alzheimer’s disease (AD). Here we show that these aggregates of phosphorylated tau protein are associated with the calcification of cell nuclei and cell projections, especially in the middle temporal, superior temporal and parahippocampal gyri. This calcification is related to the aetiology of the disease, shedding new light on the mechanisms involved in AD and potentially providing a new detection method for AD progression.


2021 ◽  
Author(s):  
Joana B. Pereira ◽  
Shorena Janelidze ◽  
Ruben Smith ◽  
Niklas Mattsson-Carlgren ◽  
Sebastian Palmqvist ◽  
...  

AbstractAlthough recent clinical trials targeting amyloid-β (Aβ) in Alzheimer’s disease (AD) have shown promising results, there is increasing evidence suggesting that understanding alternative disease pathways that interact with Aβ 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 AD. However, to this date, no studies have assessed whether astrocytosis is independently related to Aβ or tau pathology, respectively, in vivo. To address this question, we determined the levels of the astrocytic marker glial fibrillary acidic protein (GFAP) in plasma and cerebrospinal fluid (CSF) of 217 Aβ-negative cognitively unimpaired individuals, 71 Aβ-positive cognitively unimpaired individuals, 78 Aβ-positive cognitively impaired individuals, 63 Aβ-negative cognitively impaired individuals and 75 patients with a non-AD neurodegenerative disorder from the Swedish BioFINDER-2 study. Subjects underwent longitudinal Aβ (18F-flutemetamol) and tau (18F-RO948) positron emission tomography (PET) as well as cognitive testing. We found that plasma GFAP concentration was significantly increased in all Aβ-positive groups compared with subjects without Aβ pathology (p<0.01). In addition, there were significant associations between plasma GFAP with higher Aβ-PET signal in all Aβ-positive groups, but also in cognitively normal individuals with normal Aβ values (p<0.001), which remained significant after controlling for tau-PET signal. Furthermore, plasma GFAP could predict Aβ-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, sTREM2: 0.71). Although correlations were also observed between tau-PET and plasma GFAP, these were no longer significant after controlling for Aβ-PET. In contrast to plasma GFAP, CSF GFAP concentration was significantly increased in non-AD patients compared to other groups (p<0.05) and correlated with Aβ-PET only in Aβ-positive cognitively impaired individuals (p=0.005). Finally, plasma GFAP was associated with both longitudinal Aβ-PET and cognitive decline, and mediated the effect of Aβ-PET on tau-PET burden, suggesting that astrocytosis secondary to Aβ aggregation might promote tau accumulation. Altogether, these findings indicate that plasma GFAP is an early marker associated with brain Aβ pathology but not tau aggregation, even in cognitively normal individuals with a normal Aβ status. This suggests that plasma GFAP should be incorporated in current hypothetical models of AD pathogenesis and be used as a non-invasive and accessible tool to detect early astrocytosis secondary to Aβ pathology.


Sign in / Sign up

Export Citation Format

Share Document