Skip to main content

ORIGINAL RESEARCH article

Front. Aging Neurosci.
Sec. Alzheimer's Disease and Related Dementias
Volume 16 - 2024 | doi: 10.3389/fnagi.2024.1383163

In-vivo neuronal dysfunction by Aβ and tau overlaps with brain-wide inflammatory mechanisms in Alzheimer's disease Provisionally Accepted

 Lázaro Miguel S. Rodríguez1*  Ahmed Khan1 Quadri Adewale1 Gleb Bezgin1  Joseph Therriault1 Jaime Fernandez Arias1 Stijn Servaes1 Nesrine Rahmouni1  Cecile Tissot1 Jenna Stevenson1 Hongxiu Jiang1  Xiaoqian J. Chai1  Felix Carbonell2  Pedro Rosa-Neto MD PhD1  Yasser Iturria Medina1*
  • 1McGill University, Canada
  • 2Biospective Inc., Canada

The final, formatted version of the article will be published soon.

Receive an email when it is updated
You just subscribed to receive the final version of the article

The molecular mechanisms underlying neuronal dysfunction in Alzheimer's disease (AD) remain uncharacterized. Here, we identify genes, molecular pathways and cellular components associated with whole-brain dysregulation caused by amyloid-beta (Aβ) and tau deposits in the living human brain. We obtained in-vivo resting-state functional MRI (rs-fMRI), Aβ-and tau-PET for 47 cognitively unimpaired and 16 AD participants from the Translational Biomarkers in Aging and Dementia cohort. Adverse neuronal activity impacts by Aβ and tau were quantified with personalized dynamical models by fitting pathology-mediated computational signals to the participant's real rs-fMRIs. Then, we detected robust brain-wide associations between the spatial profiles of Aβ-tau impacts and gene expression in the neurotypical transcriptome (Allen Human Brain Atlas). Within the obtained distinctive signature of in-vivo neuronal dysfunction, several genes have prominent roles in microglial activation and in interactions with Aβ and tau. Moreover, cellular vulnerability estimations revealed strong association of microglial expression patterns with Aβ and tau's synergistic impact on neuronal activity (q < 0.001). These results further support the central role of the immune system and neuroinflammatory pathways in AD pathogenesis.Neuronal dysregulation by AD pathologies also associated with neurotypical synaptic and developmental processes. In addition, we identified drug candidates from the vast LINCS library to halt or reduce the observed Aβ-tau effects on neuronal activity. Top-ranked pharmacological interventions target inflammatory, cancer and cardiovascular pathways, including specific medications undergoing clinical evaluation in AD. Our findings, based on the examination of molecular-pathological-functional interactions in humans, may accelerate the process of bringing effective therapies into clinical practice.

Keywords: Alzheimer Disease, Neuronal dysfunctions and alterations, Whole-brain modeling, Transcriptomics, Amyloid - beta, Tau & phospho-Tau protein, Inflammation, Computational drug repurposing

Received: 07 Feb 2024; Accepted: 09 May 2024.

Copyright: © 2024 Rodríguez, Khan, Adewale, Bezgin, Therriault, Fernandez Arias, Servaes, Rahmouni, Tissot, Stevenson, Jiang, Chai, Carbonell, Rosa-Neto MD PhD and Iturria Medina. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

* Correspondence:
Mx. Lázaro Miguel S. Rodríguez, McGill University, Montreal, H3A 0G4, Quebec, Canada
Dr. Yasser Iturria Medina, McGill University, Montreal, H3A 0G4, Quebec, Canada