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

Front Microbiol. 2024 Jun 11:15:1412765. doi: 10.3389/fmicb.2024.1412765. eCollection 2024.

Abstract

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 (Aβ) and alpha (α)-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 the enrichment of Bilophila, a microorganism connected to cognitive impairment, long before the 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: fecal 16S rRNA seq; gut–brain–microbiome axis; intracerebral pathologies; metabolism; microbiome; neuroinflammation; prion disease; short-chain fatty acids.

Grants and funding

The author(s) declare that financial support was received for the research, authorship, and/or publication of this article. ML was supported by the Swiss National Science Foundation (Grant number 210931). MS was supported by the Stiftung Experimentelle Biomedizin. AA was supported by the Swiss National Science Foundation and a donation from Dr. Hans Salvisberg. AA and ME are supported by Driver Grant 2017DRI17 of the Swiss Personalized Health Network and of the NOMIS Foundation, the Schwyzer-Winiker Stiftung, and the Baugarten Stiftung (coordinated by the USZ Foundation, USZF27101).