P1-480: LOCUS COERULEUS SIGNAL INTENSITY IS ASSOCIATED WITH ENTORHINAL TAU PATHOLOGY AT HIGHER LEVELS OF AMYLOID BURDEN

2006 ◽  
Vol 14 (7S_Part_9) ◽  
pp. P509-P510 ◽  
Author(s):  
Heidi IL. Jacobs ◽  
Alex Becker ◽  
Kenneth Kwong ◽  
Federico d'Oleire Uquillas ◽  
Reisa A. Sperling ◽  
...  
Author(s):  
Claire J. Ciampa ◽  
Jourdan H. Parent ◽  
Theresa M. Harrison ◽  
Rebekah M. Fain ◽  
Matthew J. Betts ◽  
...  

2021 ◽  
Vol 80 (4) ◽  
pp. 325-335
Author(s):  
Sarah C Kelly ◽  
Peter T Nelson ◽  
Scott E Counts,

Abstract Locus coeruleus (LC) neurodegeneration is associated with cognitive deterioration during the transition from normal cognition to mild cognitive impairment (MCI) and Alzheimer disease (AD). However, the extent to which LC degenerative processes differentiate cognitively normal, “resilient” subjects bearing a high AD pathological burden from those with MCI or AD remains unclear. We approached this problem by quantifying the number of LC neurons and the percentage of LC neurons bearing AT8 tau pathology, TDP-43 pathology, or a marker for DNA/RNA oxidative damage, in well-characterized subjects diagnosed as normal cognition-low AD pathology (NC-LP), NC-high AD pathology (NC-HP), MCI, or mild/moderate AD. In addition, the severity of pontine arteriolosclerosis in each subject was compared across the groups. There was a trend for a step-wise ∼20% loss of LC neuron number between the NC-LP, NC-HP and MCI subjects despite a successive, significant ∼80%–100% increase in tau pathology between these groups. In contrast, increasing pontine arteriolosclerosis severity scores and LC oxidative stress burden significantly separated the NC-LP/HP and MCI/AD groups via comparative, correlation, and regression analysis. Pontine perfusion, as well as LC neuronal metabolic and redox function, may impact noradrenergic LC modulation of cognition during the preclinical and prodromal stages of AD.


2020 ◽  
Vol 16 (S4) ◽  
Author(s):  
Pauline Olivieri ◽  
Julien Lagarde ◽  
Stéphane Lehéricy ◽  
Romain Valabrègue ◽  
Fabien Caillé ◽  
...  

Neurology ◽  
2020 ◽  
Vol 94 (18) ◽  
pp. e1916-e1928
Author(s):  
Heidi I.L. Jacobs ◽  
Jean C. Augustinack ◽  
Aaron P. Schultz ◽  
Bernard J. Hanseeuw ◽  
Joseph Locascio ◽  
...  

ObjectiveTo identify the hippocampal subregions linking initial amyloid and tau pathology to memory performance in clinically normal older individuals, reflecting preclinical Alzheimer disease (AD).MethodsA total of 127 individuals from the Harvard Aging Brain Study (mean age 76.22 ± 6.42 years, 68 women [53.5%]) with a Clinical Dementia Rating score of 0, a flortaucipir tau-PET scan, a Pittsburgh compound B amyloid-PET scan, a structural MRI scan, and cognitive testing were included. From these images, we calculated neocortical, hippocampal, and entorhinal amyloid pathology; entorhinal and hippocampal tau pathology; and the volumes of 6 hippocampal subregions and total hippocampal volume. Memory was assessed with the selective reminding test. Mediation and moderation analyses modeled associations between regional markers and memory. Analyses included covariates for age, sex, and education.ResultsNeocortical amyloid, entorhinal tau, and presubiculum volume univariately associated with memory performance. The relationship between neocortical amyloid and memory was mediated by entorhinal tau and presubiculum volume, which was modified by hippocampal amyloid burden. With other biomarkers held constant, presubiculum volume was the only marker predicting memory performance in the total sample and in individuals with elevated hippocampal amyloid burden.ConclusionsThe presubiculum captures unique AD-related biological variation that is not reflected in total hippocampal volume. Presubiculum volume may be a promising marker of imminent memory problems and can contribute to understanding the interaction between incipient AD-related pathologies and memory performance. The modulation by hippocampal amyloid suggests that amyloid is a necessary, but not sufficient, process to drive neurodegeneration in memory-related regions.


2019 ◽  
Vol 15 ◽  
pp. P1113-P1114
Author(s):  
Heidi IL. Jacobs ◽  
Joyita Dutta ◽  
Alex Becker ◽  
Bernard J. Hanseeuw ◽  
Jorge Sepulcre ◽  
...  

2019 ◽  
Vol 15 ◽  
pp. P771-P772
Author(s):  
Dahyun Yi ◽  
Min Soo Byun ◽  
Jun Ho Lee ◽  
So Yeon Jeon ◽  
Gihwan Byeon ◽  
...  

2015 ◽  
Vol 130 (3) ◽  
pp. 349-362 ◽  
Author(s):  
Michiyo Iba ◽  
Jennifer D. McBride ◽  
Jing L. Guo ◽  
Bin Zhang ◽  
John Q. Trojanowski ◽  
...  

2019 ◽  
Author(s):  
Seong Su Kang ◽  
Xia Liu ◽  
Eun Hee Ahn ◽  
Jie Xiang ◽  
Fredric P. Manfredsson ◽  
...  

AbstractAberrant Tau inclusions in the locus coeruleus (LC) are the earliest detectable Alzheimer’s disease (AD)-like neuropathology in the human brain; however, why LC neurons are selectively vulnerable to developing early Tau pathology and degenerating later in disease and whether the LC might seed the stereotypical spread of Tau pathology to the rest of the brain remain unclear. Here we show that 3,4-dihydroxyphenylglycolaldehyde (DOPEGAL), which is produced exclusively in noradrenergic neurons by monoamine oxidase A (MAO-A) metabolism of norepinephrine (NE), activates asparagine endopeptidase (AEP) that cleaves Tau at residue N368 into aggregation- and propagation-prone forms, thereby leading to LC degeneration and the spread of Tau pathology. DOPEGAL triggers AEP-cleaved Tau aggregationin vitroand in intact cells, resulting in LC neurotoxicity and propagation of pathology to the forebrain. Thus, our findings reveal a novel molecular mechanism underlying the selective vulnerability of LC neurons in AD.


Sign in / Sign up

Export Citation Format

Share Document