4.7 Article

Microbial BMAA elicits mitochondrial dysfunction, innate immunity activation, and Alzheimer's disease features in cortical neurons

Journal

JOURNAL OF NEUROINFLAMMATION
Volume 17, Issue 1, Pages -

Publisher

BMC
DOI: 10.1186/s12974-020-02004-y

Keywords

Alzheimer's disease; beta-N-Methylamino-l-alanine; Mitochondrial dysfunction; Neuronal innate immunity; Neurodegeneration

Funding

  1. Faculty of Medicine, University of Coimbra [FMUC-PEPITA-2018]
  2. Santa Casa da Misericordia de Lisboa - Mantero Belard Neurosciences Prize [MB-40-2016]
  3. Fundo Europeu de Desenvolvimento Regional (FEDER) through the COMPETE 2020-Operational Programme for Competitiveness and Internationalisation (POCI)
  4. Portuguese funds through FCT-Fundacao para a Ciencia e a Tecnologia [UIBD/NEU/04539/2020, POCI-01-0145-FEDER-030712]

Ask authors/readers for more resources

BackgroundAfter decades of research recognizing it as a complex multifactorial disorder, sporadic Alzheimer's disease (sAD) still has no known etiology. Adding to the myriad of different pathways involved, bacterial neurotoxins are assuming greater importance in the etiology and/or progression of sAD. beta -N-Methylamino-l-alanine (BMAA), a neurotoxin produced by some microorganisms namely cyanobacteria, was previously detected in the brains of AD patients. Indeed, the consumption of BMAA-enriched foods has been proposed to induce amyotrophic lateral sclerosis-parkinsonism-dementia complex (ALS-PDC), which implicated this microbial metabolite in neurodegeneration mechanisms.MethodsFreshly isolated mitochondria from C57BL/6 mice were treated with BMAA and O-2 consumption rates were determined. O-2 consumption and glycolysis rates were also measured in mouse primary cortical neuronal cultures. Further, mitochondrial membrane potential and ROS production were evaluated by fluorimetry and the integrity of mitochondrial network was examined by immunofluorescence. Finally, the ability of BMAA to activate neuronal innate immunity was quantified by addressing TLRs (Toll-like receptors) expression, p65 NF-kappa B translocation into the nucleus, increased expression of NLRP3 (Nod-like receptor 3), and pro-IL-1 beta. Caspase-1 activity was evaluated using a colorimetric substrate and mature IL-1 beta levels were also determined by ELISA.ResultsTreatment with BMAA reduced O-2 consumption rates in both isolated mitochondria and in primary cortical cultures, with additional reduced glycolytic rates, decrease mitochondrial potential and increased ROS production. The mitochondrial network was found to be fragmented, which resulted in cardiolipin exposure that stimulated inflammasome NLRP3, reinforced by decreased mitochondrial turnover, as indicated by increased p62 levels. BMAA treatment also activated neuronal extracellular TLR4 and intracellular TLR3, inducing p65 NF-kappa B translocation into the nucleus and activating the transcription of NLRP3 and pro-IL-1 beta. Increased caspase-1 activity resulted in elevated levels of mature IL-1 beta. These alterations in mitochondrial metabolism and inflammation increased Tau phosphorylation and A beta peptides production, two hallmarks of AD.ConclusionsHere we propose a unifying mechanism for AD neurodegeneration in which a microbial toxin can induce mitochondrial dysfunction and activate neuronal innate immunity, which ultimately results in Tau and A beta pathology. Our data show that neurons, alone, can mount inflammatory responses, a role previously attributed exclusively to glial cells.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available