Akkermansia muciniphila identified as key strain to alleviate gut barrier injury through Wnt signaling pathway

  1. Hainan Institute of Zhejiang University, Yongyou Industry Park, Yazhou Bay Sci-Tech City, Sanya, China
  2. Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China

Peer review process

Revised: This Reviewed Preprint has been revised by the authors in response to the previous round of peer review; the eLife assessment and the public reviews have been updated where necessary by the editors and peer reviewers.

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Caetano Antunes
    University of Kansas, Lawrence, United States of America
  • Senior Editor
    Wendy Garrett
    Harvard T.H. Chan School of Public Health, Boston, United States of America

Reviewer #2 (Public Review):

Ma X. et al proposed that A. muciniphila was a key strain that promotes the proliferation and differentiation of intestinal stem cells through acting on the Wnt/b-catenin signaling pathway. They used various models, such as piglet model, mouse model and intestinal organoids to address how A. muciniphila and B. fragilis offer the protection against ETEC infection. They showed that FMT with fecal samples, A. muciniphila or B. fragilis protected piglets and/or mice from ETEC infection, and this protection is manifested as reduced intestinal inflammation/bacterial colonization, increased tight junction/Muc2 proteins, as well as proper Treg/Th17 cells. Additionally, they demonstrated that A. muciniphila protected basal-out and/or apical-out intestinal organoids against ETEC infection via Wnt signaling.

Comments on revised version:

Please add proper references to indicate the invasion of ETEC into organoids after 1 h of infection.

Reviewer #3 (Public Review):

Summary:

The manuscript by Ma et al. describes a multi-model (pig, mouse, organoid) investigation into how fecal transplants protect against E. coli infection. The authors identify A. muciniphila and B. fragilis as two important strains and characterize how these organisms impact the epithelium by modulating host signaling pathways, namely the Wnt pathway in lgr5 intestinal stem cells.

Strengths:

The strengths of this manuscript include the use of multiple model systems and follow up mechanistic investigations to understand how A. muciniphila and B. fragilis interacted with the host to impact epithelial physiology.

Weaknesses:

After an additional revision, the bioinformatics section of the methods has changed significantly from previous versions and now indicates a third sequencer was used instead: Ion S5 XL. Important parameters required to replicate analysis have still not been provided. Inspection of the SRA data indicates a mix of Illumina MiSeq and Illumina HiSeq 2500. It is now unclear which sequencing technology was used as authors have variably reported 4 different sequencers for these samples. Appropriate metadata was not provided in the SRA, although some groups may be inferred from sample names. These changing descriptions of the methodologies and ambiguity in making the data available create concerns about rigor of study and results.

Author response:

The following is the authors’ response to the previous reviews.

Public Reviews:

Reviewer #2 (Public Review):

The authors indicated that the adherence of ETEC is to intestinal epithelial cells. However, it is also possible that the majority of ETEC may reside in the intestinal mucus, particularly under in vivo infection condition. The colonization of ETEC in the jejunum and colon of piglets (Fig 2C) and in the intestines of mice (Fig S2A) does not necessarily reflect the adherence of ETEC to epithelial cells. Please verify these observations with other methods, such as immunostaining. Also, while Salmonella enterica serovar Typhimurium or Listeria monocytogenes can invade organoids within 1 hour, it is unknown if ETEC invade into organoids in this study. Clarifying this will help resolve if A. muciniphila block the adherence and/or invasion of ETEC. Please also address if A. muciniphila metabolites could prevent ETEC infection in the organoid models.

In the original manuscript, the sentence “ETEC K88 adheres to intestinal epithelial cells and induces gut inflammation (Yu et al., 2018)” in line 447 is a reference cited for the purpose of connecting the previous and the following, and it is not our result. We have deleted this sentence on line 457. Previous studies have shown that ETEC enter into intestinal epithelial cells after only one hour of infection (Xiao et al., 2022; Qian et al., 2023). Whether A. muciniphila metabolites prevent ETEC infection in the organoid models is not the focus of this manuscript, it may be further explored by other members of the research group in the future.

References:

Xiao K, Yang Y, Zhang Y, Lv QQ, Huang FF, Wang D, Zhao JC, Liu YL. 2022. Long-chain PUFA ameliorate enterotoxigenic Escherichia coli-induced intestinal inflammation and cell injury by modulating pyroptosis and necroptosis signaling pathways in porcine intestinal epithelial cells. Br. J. Nutr. 128(5):835-850.

Qian MQ, Zhou XC, Xu TT, Li M, Yang ZR, Han XY. 2023. Evaluation of Potential Probiotic Properties of Limosilactobacillus fermentum Derived from Piglet Feces and Influence on the Healthy and E. coli-Challenged Porcine Intestine. Microorganisms. 11(4).

Reviewer #3 (Public Review):

Summary:

The manuscript by Ma et al. describes a multi-model (pig, mouse, organoid) investigation into how fecal transplants protect against E. coli infection. The authors identify A. muciniphila and B. fragilis as two important strains and characterize how these organisms impact the epithelium by modulating host signaling pathways, namely the Wnt pathway in lgr5 intestinal stem cells.

Strengths:

The strengths of this manuscript include the use of multiple model systems and follow up mechanistic investigations to understand how A. muciniphila and B. fragilis interacted with the host to impact epithelial physiology.

Weaknesses:

After revision, the bioinformatics section of the methods is still jumbled and may indicate issues in the pipeline. Important parameters are not included to replicate analyses. Merging the forward and reverse reads may represent a problem for denoising. Chimera detection was performed prior to denoising.

Potential denoising issues for NovaSeq data was not addressed in the response. The authors did not clarify if multiple testing correction was applied; however, it may be assumed not as written. The raw sequencing data made available through the SRA accession (if for the correct project) indicates it was a MiSeq platform; however, the sample names do not appear to link up to this experimental design and metadata not sufficient to replicate analyses.

We have redescribed the method for microbiome sequencing analysis on lines 298-327.

Recommendations for the authors:

Reviewer #3 (Recommendations For The Authors):

SRA accession must be confirmed and metadata made available.

We updated the SRA data.

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation