Skip to main content

ORIGINAL RESEARCH article

Front. Microbiol.
Sec. Infectious Agents and Disease
Volume 15 - 2024 | doi: 10.3389/fmicb.2024.1412765
This article is part of the Research Topic Acute/chronic pathogen infections. Roles of resident, probiotic, or live biotherapeutic microbes in limiting or preventing disease View all 20 articles

Longitudinal microbiome investigation throughout prion disease course reveals pre-and clinical compositional perturbations linked to short chain fatty acid metabolism and cognitive impairment in mice

Provisionally accepted
  • University Hospital Zürich, Zurich, Switzerland

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

    Commensal intestinal bacteria shape our microbiome and have decisive roles in preserving host metabolic and immune homeostasis. They conspicuously impact disease development and progression, including amyloid-beta (A) and alpha-synuclein pathology in neurodegenerative diseases conveying the importance of the brain-gut-microbiome axis in such conditions. However, little is known about the longitudinal microbiome landscape and its potential clinical implications in other protein misfolding disorders such as prion disease. We investigated the microbiome architecture throughout prion disease course in mice. Fecal specimens were assessed by 16S ribosomal RNA sequencing. We report a temporal microbiome signature in prion disease and uncovered alterations in Lachnospiraceae, Ruminococcaceae, Desulfovibrionaceae and Muribaculaceae family members in this disease. Moreover, we determined enrichment of Bilophila, a microorganism connected to cognitive impairment, long before clinical manifestation of disease symptoms. Based on temporal microbial abundances, several associated metabolic pathways and resulting metabolites including short chain fatty acids were linked to the disease. We propose that neuroinflammatory processes relate to perturbations of the intestinal microbiome and metabolic state by an interorgan brain-gut crosstalk. Furthermore, we describe biomarkers possibly suitable for early disease diagnostics and anti-prion therapy monitoring. While our study is confined to prion disease, our discoveries might be of equivalent relevance in other proteinopathies and central nervous system pathologies.

    Keywords: Gut-brain-microbiome axis, microbiome, fecal 16S rRNA seq, Metabolism, Short Chain Fatty Acids, Prion Disease, Neuroinflammation, intracerebral pathologies

    Received: 08 Apr 2024; Accepted: 21 May 2024.

    Copyright: © 2024 Losa, Morsy, Emmenegger, Manz, Schwarz, Aguzzi and Scharl. 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: Michael Scharl, University Hospital Zürich, Zurich, Switzerland

    Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.