Peptidoglycan-Chi3l1 interaction shapes gut microbiota in intestinal mucus layer

Curation statements for this article:
  • Curated by eLife

    eLife logo

    eLife assessment

    This valuable study demonstrates that the Chitinase 3-like protein 1 (Chi3l1) interacts with gut microbiota and protects animals from intestinal injury in laboratory colitis model. The evidence supporting the claims of the authors is considered incomplete. The inclusion of consistent in vivo and in vitro data would have strengthened the study. The work will be of interest to scientists studying crosstalk between gut microbiota and inflammatory diseases.

This article has been Reviewed by the following groups

Read the full article See related articles

Abstract

The balanced gut microbiota in intestinal mucus layer plays an instrumental role in the health of the host. However, the mechanisms by which the host regulates microbial communities in the mucus layer remain largely unknown. Here, we discovered that the host regulates bacterial colonization in the gut mucus layer by producing a protein called Chitinase 3-like protein 1 (Chi3l1). Intestinal epithelial cells are stimulated by the gut microbiota to express Chi3l1. Once expressed, Chi3l1 is secreted into the mucus layer where it interacts with the gut microbiota, specifically through a component of bacterial cell walls called peptidoglycan. This interaction between Chi3l1 and bacteria is beneficial for the colonization of bacteria in the mucus, particularly for gram-positive bacteria like Lactobacillus . Moreover, a deficiency of Chi3l1 leads to an imbalance in the gut microbiota, which exacerbates colitis induced by dextran sodium sulfate (DSS). By performing fecal microbiota transplantation from Villin-cre mice or replenishing Lactobacillus in IEC ΔChil1 mice, we were able to restore their colitis to the same level as that of Villin-cre mice. In summary, this study shows a “scaffold model” for microbiota homeostasis by interaction between intestinal Chi3l1 and bacteria cell wall interaction, and it also highlights that an unbalanced gut microbiota in the intestinal mucus contributes to the development of colitis.

Article activity feed

  1. eLife assessment

    This valuable study demonstrates that the Chitinase 3-like protein 1 (Chi3l1) interacts with gut microbiota and protects animals from intestinal injury in laboratory colitis model. The evidence supporting the claims of the authors is considered incomplete. The inclusion of consistent in vivo and in vitro data would have strengthened the study. The work will be of interest to scientists studying crosstalk between gut microbiota and inflammatory diseases.

  2. Reviewer #1 (Public Review):

    The manuscript by Chen et al. investigated the interaction between CHI3L1, a chitinase-like protein in the 18 glycosyl hydrolase family, and gut bacteria in the mucosal layers. The authors provided evidence to document the direct interaction between CHI3L1 and peptidoglycan, a major component of bacterial cell walls. In doing so, Chi3l1 produced by gut epithelial cells regulates the balance of the gut microbiome and diminishes DSS-induced colitis, potentially through the colonization of protective gram-positive bacteria such as lactobacillus.

    The study is the first to systemically document the interactions between Chi3L1 and microbiome. Convincing data were shown to characterize the imbalance of gram-positive bacteria in the newly generated gut epithelial-specific Chi3L1 deficient mice. Comprehensive FMT experiments were performed to demonstrate the contributions of gut microbiome using the mouse colitis model. However, the manuscript could've been strengthened by additional mechanistic studies concerning the binding between Chi3l1 and peptidoglycan, and how this interaction could facilitate the colonization of gram-positive bacteria. Additionally, the conclusion by the authors that disordered intestinal bacteria in gut epithelial-specific Chi3L1 deficient mice, rather than an effect by host cells, contributes to exacerbated colitis, needs further validation. In fact, the fact that FMT did not completely rescue the phenotype may point to the role of host cells in the processes. On the contrary, there is an existing body of literature demonstrating the detrimental roles of Chi3l1 in the mouse IBD model, conflicting with the current study. The differences in study design and approaches in these studies that lead to controversial findings will need to be discussed.

    Specifically,

    1. In Figure 1, it is curious that the authors only chose E.coli and staphytlococcus sciuri to test the induction of Chi3l1. What about other bacteria? Why does only E.coli but not staphytlococcus sciuri induce chi3l1 production? It does not prove that the gut microbiome induces the expression of Chi3l1. If it is the effect of LPS, does it trigger a cell death response or inflammatory responses that are known to induce chi3l1 production? What is the role of peptidoglycan in this experiment? Also, it is recommended to change WT to SPF in the figure and text, as no genetic manipulation was involved in this figure.

    2. In Figure 2, the binding between Chi3l1 and PGN needs better characterization, regarding the affinity and how it compares with the binding between Chi3l1 and chitin. More importantly, it is unclear how this interaction could facilitate the colonization of gram-positive bacteria.

    3. In Figure 3, the abundance of furmicutes and other gram-positive species is lower in the knockout mice. What is the rationale for choosing lactobacillus in the following transfer experiments?

    4. FDAA-labeled E. faecalis colonization is decreased in the knockouts. Is it specific for E. faecalis, or it is generally true for all gram-positive bacteria? What about the colonization of gram-negative bacteria?

    5. In Figure 5, the fact that FMT did not completely rescue the phenotype may point to the role of host cells in the processes. The reason that lactobacillus transfer did completely rescue the phenotypes could be due to the overwhelming protective role of lactobacillus itself, as the experiments were missing villin-cre mice transferred with lactobacillus.

    6. Conflicting literature demonstrating the detrimental roles of Chi3l1 in mouse IBD model needs to be acknowledged and discussed.

  3. Reviewer #2 (Public Review):

    Chen et al. investigated the regulatory mechanism of bacterial colonization in the intestinal mucus layer in mice and its implications for intestinal diseases. They demonstrated that Chi3l1 is a protein produced and secreted by intestinal epithelial cells into the mucus layer upon response to the gut microbiota, which has a turnover effect on facilitating the colonization of gram-positive bacteria in the mucosa. The data also indicate that Chi3l1 interacts with the peptidoglycan of the bacteria cell wall, supporting the colonization of beneficial bacteria strains such as Lactobacillus, and that deficiency in Chi3l1 predisposes mice to colitis. The inclusion of a small but pertinent piece of human data added to solidify their findings in mice.

    Overall, the experiments performed were appropriate and well executed, but the data analysis is incomplete and needs to be extended. Also, additional experiments are necessary for clarification and stronger support for their conclusions.

    1. Images are of great quality but lack proper quantification and statistical analysis. Statements such as "substantial increase of Chi3l1 expression in SPF mice" (Fig.1A), "reduced levels of Firmicutes in the colon lumen of IECChil1" (Fig.3F), "Chil1-/- had much lower colonization of E.faecalis" (Fig.4G), or "deletion of Chi3l1 significantly reduced mucus layer thickness" (Supplemental Figure 3A-B) are subjective. Since many conclusions were based on imaging data, the authors must provide reliable measures for comparison between conditions, as long as possible, such as fluorescence intensity, area, density, etc, as well as plots and statistical analysis.

    2. In the fecal/Lactobacillus transplantation experiments, oral gavage of Lactobacillus to IECChil1 mice ameliorated the colitis phenotype, by preventing colon length reduction, weight loss, and colon inflammation. These findings seem to go against the notion that Chi3l1 is necessary for the colonization of Lactobacillus in the intestinal mucosa. The authors could speculate on how Lactobacillus administration is still beneficial in the absence of Chi3l1. Perhaps, additional data showing the localization of the orally administered bacteria in the gut of Chi3l1 deficient mice would clarify whether Lactobacillus are more successfully colonizing other regions of the gut, but not the mucus layer. Alternatively, later time points of 2% DSS challenge, after Lactobacillus transplantation, would suggest whether the gut colonization by Lactobacillus and therefore the milder colitis phenotype, is sustained for longer periods in the absence of Chi3l1.

  4. Reviewer #3 (Public Review):

    Summary:
    Chen et al. are addressing a fundamental question in mammalian gut biology, namely how the host controls a mutualistic host-microbiota symbiosis. The authors focus on a protein called Chitinase 3-like protein 1 (Ch3l1) and its interaction with the protective colonic mucus layer. The rationale for the study comes from previous work showing that microbial-associated molecular patterns (MAMPs) can induce Ch3l1 in vitro, but its biological functions in the colon are unknown. In this study, the authors provide evidence supporting the claim that the gut microbiota induces the expression of Ch3l1 in vivo, mainly in mucus-producing goblet cells. Insightfully, the authors note that Ch3l1, although it lacks enzyme (chitinase) activity, still binds Chitin, a glycan that has structural similarity to bacterial cell wall peptidoglycan. This leads the authors to hypothesize that Ch3l1 binds microbial cell walls, particularly those of peptidoglycan-rich Gram-positive probiotic bacteria within the mucus, to promote their retention in the colon. Using a combination of in vivo work with mice conditionally lacking Ch3l1 in gut epithelium (IEC Ch3l1 KO); microbiota profiling; imaging of host-microbiota interactions with labeled microbes; and fecal transplants, the authors provide compelling evidence that Ch3l1 is secreted into the gut mucus layer and that the presence of Ch3l1 is associated with increased levels of beneficial Gram+ bacteria, including Lactobacillus spp. In turn, using a well-characterized colitis model, the authors show that Ch3l1 is associated with protection from intestinal injury caused by Dextran Sodium Sulfate. While these studies are novel and informative, there are several issues that undermine the authors' conclusions.

    Strengths:
    The authors nicely link microbial induction of Ch3l1 to mucosal protection from intestinal injury. This is done through the use of germ-free and ex-germ-free studies and by comparing Ch3l1 expression in situ between them; microbial sequencing between Control and IEC Ch3l1 KO mice, and clinical and histological injury metrics between these strains. The authors convincingly demonstrate the presence of Ch3l1 in the gut mucus through imaging, and that the deletion of this protein in mice alters the microbiota by reducing the relative abundance of Gram-positive species.

    The study employs a technically diverse set of analyses to address their hypothesis, including fluorescent labelling of microbial species for add-back studies, fecal transplants to distinguish the role(s) of the microbiota vs. host in the IEC Ch3l1 KO phenotypes in the intestinal challenge models.

    Weaknesses:
    The claim that mucus-associated Ch3l1 controls colonization of beneficial Gram-positive species within the mucus is not conclusive. The study should take into account recent discoveries on the nature of mucus in the colon, namely its mobile fecal association and complex structure based on two distinct mucus barrier layers coming from proximal and distal parts of the colon (PMID: ). This impacts the interpretation of how and where Ch3l1 is expressed and gets into the mucus to promote colonization. It also impacts their conclusions because the authors compare fecal vs. tissue mucus, but most of the mucus would be attached to the feces. Of the mucus that was claimed to be isolated from the WT and IEC Ch3l1 KO, this was not biochemically verified. Such verification (e.g. through Western blot) would increase confidence in the data presented. Further, the study relies upon relative microbial profiling, which can mask absolute numbers, making the claim of reduced overall Gram-positive species in mice lacking Ch3l1 unproven. It would be beneficial to show more quantitative approaches (e.g. Quantitative Microbial Profiling, QMP) to provide more definitive conclusions on the impact of Ch3l1 loss on Gram+ microbes.

    Other weaknesses lie in the execution of the aims, leaving many claims incompletely substantiated. For example, much of the imaging data is challenging for the reader to interpret due to it being unfocused, too low of magnification, not including the correct control, and not comparing the same regions of tissues among different in vivo study groups. Statistical rigor could be better demonstrated, particularly when making claims based on imaging data. These are often presented as single images without any statistics (i.e. analysis of multiple images and biological replicates). These images include the LTA signal differences, FISH images, Enterococcus colonization, and mucus thickness.