New Paper: Metaproteomic Profiling of Fungal Gut Colonization in Gnotobiotic Mice

Congratulations to our Platform 2 Co-Lead Dr. Marie-Claire Arrieta on this recent publication!

Figure 1. Description of a gnotobiotic colonisation model by label-free quantitative metaproteomics. A Experimental design of shotgun label-free proteomic experiment, including enrichment of fecal microbial cells by differential centrifugation. B Detected fecal metaproteome composed of proteins originating from 12 bacterial, 5 fungal species, and mouse. C PCA score plot based on the relative levels of 3845 proteins quantified for mouse, bacteria, and fungi/yeast. X- and Y-axis show the first and second principal components, accounting for 53.1% and 12.2% of the total variation, respectively. Abbreviations of the mice treatment groups: B, bacteria; BY, bacteria-yeast; BY_ABX/AFX, bacteria-yeast and antibiotic or antifungal treatment; GF, germ-free; Y, fungi/yeast

Abstract

Eukaryotic microbes can modulate mammalian host health and disease states, yet the molecular contribution of gut fungi remains nascent. We previously showed that mice exclusively colonised with fungi displayed increased sensitivity to allergic airway inflammation and had fecal metabolite profiles similar to germ-free mice. This marginal effect on the host metabolome suggested that fungi do not primarily use metabolites to modulate the host immune system.

To describe functional changes attributed to fungal colonisation, we performed mass spectrometry-based analyses of feces (Label-Free Quantitative; LFQ) and the small intestine (labeling with Tandem Mass Tag; TMT) of gnotobiotic mice colonised with defined consortia of twelve bacterial species, five fungal species, or both. We also evaluated the effect of microbiome perturbances on the metaproteome by analysing feces from mouse pups treated with an antibiotic or antifungal.

We detected 6675 proteins in the mice feces, of which 3845 had determined LFQ levels. Analysis of variance showed changes in the different gnotobiotic mouse groups; specifically, 46% of 2860 bacterial, 15% of 580 fungal, and 76% of 405 mouse quantified proteins displayed differential levels. The antimicrobial treatments resulted in lasting changes in the bacterial and fungal proteomes, suggesting that the antimicrobials impacted the entire community. Fungal colonisation resulted in changes in host proteins functional in innate immunity as well as metabolism, predicting specific roles of gut fungi on host systems during early developmental stages. Several of the detected fungal proteins (3% of 1492) have been previously reported as part of extracellular vesicles and having immunomodulating properties. Using an isobaric labelling TMT approach for profiling low abundant proteins of the jejunal tissue, we confirmed that the five fungal species differentially impacted the host intestinal proteome compared to the bacterial consortium. The detected changes in mouse jejunal proteins (4% of 1514) were mainly driven by metabolic proteins.

We used quantitative proteomic profiling of gnotobiotic conditions to show how colonisation with selected fungal species impacts the host gut proteome. Our results suggest that an increased abundance of certain gut fungal species in early life may affect the developing intracellular attributes of epithelial and immune cells.

Publication: Metaproteomic profiling of fungal gut colonization in gnotobiotic mice. Pettersen VK, Dufour A, Arrieta AC. Animal Microbiome. 22 Feb 2022.