1,563
Views
1
CrossRef citations to date
0
Altmetric
Research Paper

Alterations in fecal β-defensin-3 secretion as a marker of instability of the gut microbiota

, , , , & ORCID Icon
Article: 2233679 | Received 20 Feb 2023, Accepted 03 Jul 2023, Published online: 18 Jul 2023

ABSTRACT

Compositional changes in the microbiota (dysbiosis) may be a basis for Irritable Bowel Syndrome (IBS), but biomarkers are currently unavailable to direct microbiota-directed therapy. We therefore examined whether changes in fecal β-defensin could be a marker of dysbiosis in a murine model. Experimental dysbiosis was induced using four interventions relevant to IBS: a mix of antimicrobials, westernized diets (high-fat/high-sugar and high salt diets), or mild restraint stress. Fecal mouse β-defensin-3 and 16S rRNA-based microbiome profiles were assessed at baseline and during and following these interventions. Each intervention, except for mild restraint stress, altered compositional and diversity profiles of the microbiota. Exposure to antimicrobials or a high-fat/high-sugar diet, but not mild restraint stress, resulted in decreased fecal β-defensin-3 compared to baseline. In contrast, exposure to the high salt diet increased β-defensin-3 compared to baseline. Mice exposed to the mix of antimicrobials showed the largest compositional changes and the most significant correlations between β-defensin-3 levels and bacterial diversity. The high salt diet was also associated with significant correlations between changes in β-defensin-3 and bacterial diversity, and this was not accompanied by discernible inflammatory changes in the host. Thus, dietary change or antimicrobial exposure, both recognized factors in IBS exacerbations, induced marked dysbiosis that was accompanied by changes in fecal β-defensin-3 levels. We propose that serial monitoring of fecal β-defensins may serve as a marker of dysbiosis and help identify those IBS patients who may benefit from microbiota-directed therapeutic interventions.

Introduction

The term dysbiosis is used here to describe changes in the microbiota composition, function or temporal stability and is thought to contribute to functional and inflammatory conditions of the gut. The gut microbial composition is influenced by both host factors including genetics, psychological stress, immune activation and altered physiology, and environmental factors such as infection, diet and antibiotic exposure.Citation1–3 Each of these factors has been implicated in the development or exacerbation of IBS. Enteric infection or extensive antibiotic exposure is the recognized risk factor for the development of IBS,Citation4,Citation5 and diet is a common trigger of symptoms in IBS.Citation6–8 These observations rationalize the choice of interventions such as antibiotic exposure, dietary change, and stress to induce dysbiosis in the present study. Although, the intestinal microbiota is stable in healthy adult humans,Citation9,Citation10 the intra-subject variation in microbiota composition is less than that seen between individuals.Citation11–14 Moreover, temporal instability of the human microbiota has been documented in some IBS patients.Citation15–17 It follows that a simple marker of dysbiosis that reflects compositional instability of gut microbiotaCitation18 would be useful in identifying those IBS patients who might benefit from microbiota-directed therapies. Interestingly, a recent studyCitation19 showed that colonization of germ-free mice with IBS microbiota induced an IBS-like phenotype in the recipient mice, strongly implying that the microbiota contributes to the expression of IBS, and this was accompanied by an increase in β-defensin-3.

Defensins are small cationic antimicrobial peptides that play a role in maintaining homeostasis between the host and its resident microbes and protecting against pathogens.Citation20 Defensins are produced mainly by colonic epithelial cells, Paneth cells and immunocytes in response to inflammatory stimuli or exogenous microbial substances,Citation21,Citation22 and serve as an effective barrier against the entry of pathogens into the host mucosa.Citation23–25 Alpha and some β-defensins are constitutively expressed within the GI tract and most remain stable during inflammation.Citation26,Citation27 However, others like β-defensin-3 (a murine homologue of human β-defensin-2) are induced primarily upon pathogen encounter.Citation20,Citation28,Citation29 Interestingly, defensins and other antimicrobial peptides also influence microbiota composition. For example, dysfunctional Paneth cells result in significant changes in microbiota composition.Citation30–32

Changes in defensin secretion have been identified in IBS. Langhorst et al.Citation33 revealed increased levels of human β-defensin-2 in the feces of IBS patients in the absence of macroscopic inflammation, as levels of fecal calprotectin and lactoferrin did not differ between healthy controls and patients with IBS. Similarly, increased concentrations of fecal β-defensin-2 were found in children with IBS, associated with abdominal pain and increased gut permeability, indicating activation of the innate immune system.Citation34 However, decreased levels of β-defensin-2 have also been reported in patients with IBS compared to controls.Citation35 A very recent study proposed that measurement of human β-defensin-2 levels, together with the inclusion of microbiome profiles, can serve as a marker to differentiate between IBD and IBS patients.Citation36 Recently, we have shown that colonizing germ-free mice with IBS-D microbiota results in altered gut function, including faster gastrointestinal transit, altered gut permeability and secretion, which was accompanied by an increase in colonic β-defensin-3 expression in recipient mice.Citation19 Together, these observations prompted the current proof-of-concept study in mice to determine whether the experimental perturbation of the intestinal microbiota, using interventions relevant to the natural history of IBS, will result in changes in fecal β-defensin-3 levels.

Results

Antimicrobials disrupt gut microbiota composition and decrease fecal β-defensin secretion in mice

A previously described mix of non-absorbable antimicrobials (AMC)Citation37,Citation38 caused a marked and persistent disruption of microbiota composition, as illustrated in the PCoA plot created using the Bray Curtis and Aitchison distance matrices (, , Supplementary Table S1). The stool microbiota profiles of the mice at baseline clustered together with a major drift following the AMC intervention and minimal reversal to baseline during the recovery period as shown in . Fecal microbial diversity did not revert to baseline one-week post-AMC as shown by the Shannon Diversity Index (). Alpha diversity was lower during the intervention period as compared to the baseline. No statistically significant changes in net bacterial load were found in mice (Supplementary Figure S2a and S2b). To confirm the impact of AMC on net microbial composition, we performed ANCOM-BC analysis to find differentially expressed bacteria. We found altered relative abundance of several bacteria taxa that persisted during recovery, and these included the genera Anaerotruncus, Butyricicoccus, Candidatus Arthromitus, Candidatus Saccharimonas, some species of Clostridium, Defluviitaleaceae, Enterorhabdus, Lachnospira, Mucispirillum, Muribaculum, Peptococcus, Prevotellaceae, Sporobacter, and Tyzzerella, among others (). While most of these bacteria were decreased during AMC administration, the abundance of the genera Bacteroides and Proteus increased in mice. AMC administration was accompanied by a decrease in β-defensin-3 levels (), which persisted during the recovery period (). There was a positive correlation between β-defensin-3 levels and overall bacterial diversity (), suggesting a linkage between these responses. Although no major differences were observed in the microbial composition and β-defensin-3 levels between males and females, subtle differences were noted. For example, while the stool microbiota profiles and bacterial taxa of all the males and females followed similar trends at baseline and during the intervention and recovery periods (Supplementary Figure S1a), only Enterococcus spp., Escherichia spp., and Shigella were increased in females during the intervention (Supplementary Figure S4). In addition, while a three-fold reduction in β-defensin-3 levels from baseline was observed in males, only over a one-fold decrease was seen in females during the AMC intervention (Supplementary Figure S1b).

Figure 1. The antimicrobial cocktail alters gut microbiota composition and induces changes in fecal β-defensin secretion in mice. a) PCoA plot based on a Bray Curtis distance matrix analysis of the fecal microbiota composition over the course of the experiment in mice. Community composition was compared by ADONIS2 with 999 permutations. Each dot represents the bacterial microbiota of an individual mouse. Ellipses indicate 95% confidence intervals and were generated as a distribution around a centroid for each group based on ADONIS2. b) Changes in alpha-diversity using Shannon diversity index, analysis was performed by 1- way ANOVA followed by Dunn’s testing for multiple comparisons. Data is presented as box and whisker plot with whiskers extending from 10th to 90th percentile. c) Heatmap of all the differentially abundant taxa (aggregated at genus level) in mice evaluated using ANCOM-BC. Statistical significance on differentially abundant taxa between groups was corrected for multiple comparisons using FDR (q < 0.05). Color intensity on heatmaps was generated by using z-scores calculated based on relative abundance of each bacterial taxa. d) Time-course graph showing fluctuations in fecal β-defensin-3 values in mice over the course of the experiment. Time points chosen for microbiota analysis are indicated with arrows. e) Composite of mean changes in β-defensin secretion during the three experimental timepoints, presented as a box and whisker plot with whiskers extending from 10th to 90th percentile. Statistical differences were calculated using the Kruskal–Wallis test followed by post hoc Dunn’s test for multiple comparisons. f) Correlation between changes in alpha diversity values and defensin secretion in mice during the course of the experiment. Red symbols represent associations between baseline and β-defensin-3 levels, while green and blue symbols represent intervention timepoints and day 21 of recovery time point, respectively. Correlations were generated using Spearman rank index. Baseline days 1 and 7 were combined for the analysis. n = 4–5 mice/group. p < 0.05 was considered significant. **** = p ≤0.0001, *** = p ≤0.001, ** = p ≤0.01, * = p ≤0.05.

Figure 1. The antimicrobial cocktail alters gut microbiota composition and induces changes in fecal β-defensin secretion in mice. a) PCoA plot based on a Bray Curtis distance matrix analysis of the fecal microbiota composition over the course of the experiment in mice. Community composition was compared by ADONIS2 with 999 permutations. Each dot represents the bacterial microbiota of an individual mouse. Ellipses indicate 95% confidence intervals and were generated as a distribution around a centroid for each group based on ADONIS2. b) Changes in alpha-diversity using Shannon diversity index, analysis was performed by 1- way ANOVA followed by Dunn’s testing for multiple comparisons. Data is presented as box and whisker plot with whiskers extending from 10th to 90th percentile. c) Heatmap of all the differentially abundant taxa (aggregated at genus level) in mice evaluated using ANCOM-BC. Statistical significance on differentially abundant taxa between groups was corrected for multiple comparisons using FDR (q < 0.05). Color intensity on heatmaps was generated by using z-scores calculated based on relative abundance of each bacterial taxa. d) Time-course graph showing fluctuations in fecal β-defensin-3 values in mice over the course of the experiment. Time points chosen for microbiota analysis are indicated with arrows. e) Composite of mean changes in β-defensin secretion during the three experimental timepoints, presented as a box and whisker plot with whiskers extending from 10th to 90th percentile. Statistical differences were calculated using the Kruskal–Wallis test followed by post hoc Dunn’s test for multiple comparisons. f) Correlation between changes in alpha diversity values and defensin secretion in mice during the course of the experiment. Red symbols represent associations between baseline and β-defensin-3 levels, while green and blue symbols represent intervention timepoints and day 21 of recovery time point, respectively. Correlations were generated using Spearman rank index. Baseline days 1 and 7 were combined for the analysis. n = 4–5 mice/group. p < 0.05 was considered significant. **** = p ≤0.0001, *** = p ≤0.001, ** = p ≤0.01, * = p ≤0.05.

Table 1. P and R2 (group variance) values based on the ADONIS2 statistical analysis of the Bray Curtis-distance matrix (top) and Aitchison distance matrix (bottom) on the effect of antimicrobials, high-fat/high-sugar diet and high salt diet on fecal microbiota composition in mice.

We next explored the relationships between bacterial taxa that were found to be differentially abundant based on ANCOM-BC analysis (), β-defensin-3 levels and the Shannon diversity index. We identified several genera that correlated with not only β-defensin-3 levels but also Shannon diversity (). These included Acetatifactor, Alistipes, Anaerotruncus, Anaerosporobacter, Bacteroides, Butyricicoccus, Candidatus Arthromitus, Candidatus_Saccharimona, Defluviitaleaceae, Enterorhabdus, Prevotellaceae, Lachnospira, Lactobacillus, Mucispirillium, Peptococcus, and Sporobacter. Interestingly, bacteria possessing protective/anti-inflammatory properties correlated negatively with β-defensin-3 levels, suggesting that low β-defensin-3 may be due to the abundance of these counter-inflammatory bacteria such as Bacteroides (r = −0.6, p < 0.001) and Alistipes (r = −0.5, p = 0.01),Citation39,Citation40 as shown in . Conversely, the abundance of bacteria with pro-inflammatory properties correlated positively with low defensin levels (). For example, Prevotellaceae with reported pro-inflammatory propertiesCitation41,Citation42 (r = 0.6, p < 0.001), Candidatus ArthromitusCitation43 (r = 0.8, p < 0.001), EisenbergiellaCitation44 (r = 0.5, p = 0.01), and Muribaculum taxaCitation45 (r = 0.6, p < 0.001) were all positively correlated with low defensin levels following AMC exposure.

Table 2. Correlations between the abundances of differentially abundant bacterial taxa (genus), defensin levels and microbial diversity (Shannon Diversity). Bacterial taxa that were found to be differentially abundant in ANCOM-BC analysis were correlated with β-defensin levels for all experimental conditions. Additional correlations were made to see whether the differentially abundant taxa that correlate with β-defensins also correlate with microbial diversity (Shannon index values were used). All correlations were based on relative abundances and were generated using the Spearman rank index followed by FDR corrections. p < 0.05 was considered significant following FDR correction.

High-fat/high-sugar diet transiently alters gut microbiota composition and decreases fecal β-defensin secretion in mice

The high-fat/high-sugar “Western” diet (HFHSD) altered bacterial composition compared to the baseline () as illustrated by beta and alpha diversity analyses, and these changes normalized within 1 week of discontinuation of the diet. The changes in beta diversity were confirmed by statistical analyses based on Bray Curtis and Aitchison Distance matrices; both matrices revealed similar results (). While no changes in net bacterial load were found in mice (Supplementary Figure S2c and S2d), changes to the fecal microbial composition were evident at genus level (). The heatmap based on ANCOM-BC analysis revealed that most of the bacterial genera that showed proportional shifts during the exposure to the diet reverted to baseline levels when the diet was discontinued in all mice (). Many bacterial taxa including Anaerosporobacter, Candidatus Arthromitus, Eisenbergiella, Muribaculum, Prevotellaceae, and Parasutterella were lower during the intervention, while taxa like Anaerotruncus, Angelakisella, Lactococcus, Ruminiclostridium, and Tyzzerella were transiently increased. As shown in , β-defensin-3 levels decreased during the diet and began to normalize during the recovery period, although they did not return to baseline levels. In addition, fecal β-defensin-3 levels positively correlated with changes in alpha diversity in mice (r = 0.6, p = 0.002) (). Interestingly, the abundance of bacteria with pro-inflammatory properties, Candidatus ArthromitusCitation43 (r = 0.5, p = 0.04) and EisenbergiellaCitation44 (r = 0.6, p < 0.001) correlated positively with low defensin levels and Shannon diversity following HFHSD exposure ().

Figure 2. The high-fat/high-sugar diet administration transiently disrupts gut microbiota composition and alters fecal β-defensin secretion in mice. a) PCoA plot based on a Bray Curtis distance matrix analysis of the fecal microbiota composition over the course of the experiment in mice. Community composition was compared by ADONIS2 with 999 permutations. Ellipses indicate 95% confidence intervals and were generated as a distribution around a centroid for each group based on ADONIS2. b) Changes in alpha-diversity using Shannon diversity index, analysis was performed by 1- way ANOVA followed by Dunn’s testing for multiple comparisons. Data is presented as box and whisker plot with whiskers extending from 10th to 90th percentile. c) Heatmap of all the significantly differential abundant taxa (aggregated at genus level) in mice evaluated using ANCOM-BC. Color intensity on heatmaps was generated by using z-scores calculated based on relative abundance of each bacterial taxa. d) Time-course graph showing fluctuations in fecal β-defensin-3 values in mice over the course of the experiment. Time points chosen for microbiota analysis are indicated with arrows. e) Composite of mean changes in β-defensin-3 secretion during the three experimental timepoints, presented as box and whisker plot with whiskers extending from 10th to 90th percentile. Statistical differences were calculated using the Kruskal–Wallis test followed by post hoc Dunn’s test for multiple comparisons. f) Correlation between changes in alpha diversity values and defensin secretion in mice during the course of the experiment. Red symbols represent associations between baseline and β-defensin levels, while green and blue symbols represent intervention timepoints and day 21 of recovery time point, respectively. Correlations were generated using Spearman rank index. Baseline days 1 and 7 were combined for the analysis. n = 4–5 mice/group. p < 0.05 was considered significant. **** = p ≤0.0001, *** = p ≤0.001, ** = p ≤0.01, * = p ≤0.05.

Figure 2. The high-fat/high-sugar diet administration transiently disrupts gut microbiota composition and alters fecal β-defensin secretion in mice. a) PCoA plot based on a Bray Curtis distance matrix analysis of the fecal microbiota composition over the course of the experiment in mice. Community composition was compared by ADONIS2 with 999 permutations. Ellipses indicate 95% confidence intervals and were generated as a distribution around a centroid for each group based on ADONIS2. b) Changes in alpha-diversity using Shannon diversity index, analysis was performed by 1- way ANOVA followed by Dunn’s testing for multiple comparisons. Data is presented as box and whisker plot with whiskers extending from 10th to 90th percentile. c) Heatmap of all the significantly differential abundant taxa (aggregated at genus level) in mice evaluated using ANCOM-BC. Color intensity on heatmaps was generated by using z-scores calculated based on relative abundance of each bacterial taxa. d) Time-course graph showing fluctuations in fecal β-defensin-3 values in mice over the course of the experiment. Time points chosen for microbiota analysis are indicated with arrows. e) Composite of mean changes in β-defensin-3 secretion during the three experimental timepoints, presented as box and whisker plot with whiskers extending from 10th to 90th percentile. Statistical differences were calculated using the Kruskal–Wallis test followed by post hoc Dunn’s test for multiple comparisons. f) Correlation between changes in alpha diversity values and defensin secretion in mice during the course of the experiment. Red symbols represent associations between baseline and β-defensin levels, while green and blue symbols represent intervention timepoints and day 21 of recovery time point, respectively. Correlations were generated using Spearman rank index. Baseline days 1 and 7 were combined for the analysis. n = 4–5 mice/group. p < 0.05 was considered significant. **** = p ≤0.0001, *** = p ≤0.001, ** = p ≤0.01, * = p ≤0.05.

As in the AMC group, in the HFHSD group, we found small differences between males and females in microbial composition and β-defensin-3 levels. For example, both sexes showed similar trends in alterations in bacterial diversity (Supplementary Figure S1c) as well as proportional shifts in bacterial genera before, during, and following the diet intervention (Supplementary Figure S5). While small differences in bacterial taxa abundances and reduced β-defensin-3 were found in both males and females, the magnitude of this change was greater in females than males, and β-defensin-3 levels normalized only in males post intervention (Supplementary Figure S1d). Specifically, defensin secretion was decreased from baseline levels by > 1-fold in males and > 2-fold in females (Supplementary Figure S1d). This difference might be due to the dissimilarity of microbiota profiles between males and females (Supplementary Figure S1c).

High salt diet alters fecal microbiota composition and increases fecal β-defensin secretion in mice

Administration of a high salt diet (HSD) also induced changes in the microbiota composition, as reflected by alpha and beta diversity analyses ( ). Statistical analyses evaluating both Bray Curtis and Aitchison distance matrices showed a dissimilarity between the intervention and baseline or recovery timepoints as the centroids were distantly grouped. The direction of changes in alpha diversity following a HSD was different from that seen following AMC and HFHSD interventions (). ANCOM-BC analysis of microbial composition revealed a reduced abundance of several microbial taxa including Lactobacillus and Candidatus Arthromitus () and increased relative abundance of Parabacteroides, Akkermansia, Anaerotruncus, Staphylococcus and Erysipelatoclostridum. In contrast to AMC and HFHSD interventions, HSD transiently increased fecal β-defensin-3 secretion up to 2-fold compared to baseline levels in all mice (; Supplementary Figure S1f). A positive correlation was found between altered Shannon diversity index values and β-defensin-3 levels (r = 0.5; p = 0.006) as shown in . Although, there was an overlap between the ellipses representing the three timepoints ( and Supplementary Figure S1e), the net compositional change based on p-values in both males and females showed dissimilarity in beta diversity. As shown in , the high salt diet decreased Lactobacillus and Candidatus Arthromitus relative abundance and increased relative abundance of Parabacteroides, Akkermansia, Anaerotruncus, Staphylococcus and Harryflintia in both males and females (Supplementary Figure S6).

Figure 3. The high salt diet alters gut microbiota composition and modulates host fecal β-defensin secretion in mice. a) PCoA plot based on a Bray Curtis distance matrix analysis of the fecal microbiota composition over the course of the experiment in mice. Community composition was compared by ADONIS2 with 999 permutations. Ellipses indicate 95% confidence intervals and were generated as a distribution around a centroid for each group based on ADONIS2. b) Changes in alpha-diversity using Shannon diversity index, analysis was performed by 1- way ANOVA followed by Dunn’s testing for multiple comparisons. Data is presented as box and whisker plot with whiskers extending from 10th to 90th percentile. c) Heatmap of all the significantly differential abundant taxa (aggregated at genus level) in mice evaluated using ANCOM-BC. Color intensity on heatmaps was generated by using z-scores calculated based on relative abundance of each bacterial taxa. d) Time-course graph showing fluctuations in fecal β-defensin-3 values in mice over the course of the experiment. Time points chosen for microbiota analysis are indicated with arrows. e) Composite of mean changes in β-defensin-3 secretion during the three experimental timepoints, presented as box and whisker plot with whiskers extending from 10th to 90th percentile. Statistical differences were calculated using the Kruskal–Wallis test followed by post hoc Dunn’s test for multiple comparisons. f) Correlation between changes in alpha diversity values and defensin secretion in mice during the course of the experiment. Red symbols represent associations between baseline and β-defensin levels, while green and blue symbols represent intervention timepoints and day 21 of recovery time point, respectively. Correlations were generated using Spearman rank index. Baseline days 1 and 7 were combined for the analysis. n = 4–5 mice/group. p < 0.05 was considered significant. **** = p ≤0.0001, *** = p ≤0.001, ** = p ≤0.01, * = p ≤0.05.

Figure 3. The high salt diet alters gut microbiota composition and modulates host fecal β-defensin secretion in mice. a) PCoA plot based on a Bray Curtis distance matrix analysis of the fecal microbiota composition over the course of the experiment in mice. Community composition was compared by ADONIS2 with 999 permutations. Ellipses indicate 95% confidence intervals and were generated as a distribution around a centroid for each group based on ADONIS2. b) Changes in alpha-diversity using Shannon diversity index, analysis was performed by 1- way ANOVA followed by Dunn’s testing for multiple comparisons. Data is presented as box and whisker plot with whiskers extending from 10th to 90th percentile. c) Heatmap of all the significantly differential abundant taxa (aggregated at genus level) in mice evaluated using ANCOM-BC. Color intensity on heatmaps was generated by using z-scores calculated based on relative abundance of each bacterial taxa. d) Time-course graph showing fluctuations in fecal β-defensin-3 values in mice over the course of the experiment. Time points chosen for microbiota analysis are indicated with arrows. e) Composite of mean changes in β-defensin-3 secretion during the three experimental timepoints, presented as box and whisker plot with whiskers extending from 10th to 90th percentile. Statistical differences were calculated using the Kruskal–Wallis test followed by post hoc Dunn’s test for multiple comparisons. f) Correlation between changes in alpha diversity values and defensin secretion in mice during the course of the experiment. Red symbols represent associations between baseline and β-defensin levels, while green and blue symbols represent intervention timepoints and day 21 of recovery time point, respectively. Correlations were generated using Spearman rank index. Baseline days 1 and 7 were combined for the analysis. n = 4–5 mice/group. p < 0.05 was considered significant. **** = p ≤0.0001, *** = p ≤0.001, ** = p ≤0.01, * = p ≤0.05.

Interestingly, the abundance of bacteria with pro-inflammatory properties correlated positively with high defensin levels, suggesting that high β-defensin-3 may be due to the presence of these pro-inflammatory bacteria. For example, EisenbergiellaCitation44 (r = 0.5, p = 0.02) positively correlated with high defensin levels following HSD exposure, as shown in . Conversely, bacteria possessing protective/anti-inflammatory properties such as Clostridium_sensu_stricto_1Citation46 (r = −0.46, p = 0.04) and LactobacillusCitation47 (r = −0.7, p < 0.001) correlated negatively with β-defensin-3 levels.

The effect of high salt diet on β-defensin secretion is not mediated by overt inflammation or immune activation

Previous work from our laboratory showed that high salt diet administration for 4 weeks induced a dysbiosis that was associated with an increased susceptibility to colitis in mice.Citation48 We questioned whether the increase in β-defensin-3 levels following high salt diet was a component of a diet-induced inflammatory response. We therefore administered a high salt or control diet to mice for one-week and assessed myeloperoxidase (MPO) activity, and inflammation-associated gene expression in the colon. We found no changes in MPO activity in mice following the diet (). As shown in , of the 68 genes related to gut immune and barrier function, T-bet, responsible for regulating intestinal inflammation,Citation49 was found to be differentially expressed in control vs. high salt diet treated mice (log2ratio = 1.31; p = 0.0081).

Figure 4. The administration of high salt diet does not induce overt inflammation in mice. a) Colonic myeloperoxidase (MPO) levels in mice who received the control diet, the high salt diet or were given high salt diet followed by 7-day recovery period. Statistical differences between groups were evaluated using 1-way ANOVA (non-parametric) followed by post-hoc Dunn’s analysis. Values are presented as box and whiskers plot with whiskers extending from 10th to 90th percentile, n = 5 mice/control and recovery groups; and n= 4 mice/treatment group with p < 0.05 considered as significant. b) Volcano plot of gene expression in colonic tissues of mice receiving high salt diet or control diet for 1 week. Red dot represents gene with p < 0.01. Black dots are genes with 0.01 > p > 0.05. Genes present on the right side of the zero were upregulated in high salt diet treated mice but downregulated in control mice. The heat map summarizes the expression of T-bet gene associated with innate immunity in the colon tissues of mice treated with high salt diet or a control diet. Red represents the samples with genes that were downregulated, while green represents the samples with genes that were upregulated. n = 4 mice/group. T-bet: T-box transcription factor.

Figure 4. The administration of high salt diet does not induce overt inflammation in mice. a) Colonic myeloperoxidase (MPO) levels in mice who received the control diet, the high salt diet or were given high salt diet followed by 7-day recovery period. Statistical differences between groups were evaluated using 1-way ANOVA (non-parametric) followed by post-hoc Dunn’s analysis. Values are presented as box and whiskers plot with whiskers extending from 10th to 90th percentile, n = 5 mice/control and recovery groups; and n= 4 mice/treatment group with p < 0.05 considered as significant. b) Volcano plot of gene expression in colonic tissues of mice receiving high salt diet or control diet for 1 week. Red dot represents gene with p < 0.01. Black dots are genes with 0.01 > p > 0.05. Genes present on the right side of the zero were upregulated in high salt diet treated mice but downregulated in control mice. The heat map summarizes the expression of T-bet gene associated with innate immunity in the colon tissues of mice treated with high salt diet or a control diet. Red represents the samples with genes that were downregulated, while green represents the samples with genes that were upregulated. n = 4 mice/group. T-bet: T-box transcription factor.

Mild repeated restraint stress does not alter gut microbiota composition or host fecal β-defensin secretion in mice

The effect of mild repeated stress on the microbial composition was negligible, compared to unstressed mice (Supplementary Figure S3a). Similarly, there were no differences in β-defensin-3 secretion in stressed vs. control mice (Supplementary Figure S3b and S3c) or between males and females (Supplementary Figure S3d). No correlations were found between β-defensin-3 levels and overall bacterial diversity (Supplementary Figure S3e).

Discussion

Our findings indicate that experimentally induced changes in gut microbiota composition result in increases or decreases in β-defensin-3 secretion. Previous studies on β-defensin levels in IBS patients or relevant mouse models were based on samples collected at single timepoints,Citation33,Citation35,Citation36 whereas our study involved several time points over 7 days. Given that our interventions produced either increases or decreases in β-defensin-3 compared to baseline, single time point measurements may not capture these changes.

It follows that serial measurements of fecal β-defensins may better capture dysbiosis in IBS patients.Citation50 The relationship of innate immune activation, altered microbiota composition and the changes in gut physiology that underlie symptom generation in IBS is complex.Citation51 Changes in physiology alter the microbial habitat and thus microbiota composition and, conversely, dysbiosis influences gut physiology, resulting in long-term instability of the host-microbial relationship. Extrinsic factors including antimicrobial exposure or dietary change further influence microbial composition. Based on the results of our study, we propose that variation in β-defensin secretion is secondary to altered microbiota composition and that monitoring β-defensins is a putative biomarker of dysbiosis in IBS. Such a biomarker would help identify those patients in whom microbiota-directed therapies are likely to offer more therapeutic benefit. Current controversies regarding the efficacy of therapies such as probiotics and fecal microbial transfer may simply reflect the fact that while these interventions were aimed at patients exhibiting symptomatic homogeneity, not all patients exhibit,Citation50 thereby diluting the overall therapeutic benefit of microbiota-directed therapy.

Previous studies on inducible β-defensins in IBSCitation33 and related disorders like Inflammatory Bowel DiseaseCitation52,Citation53 have proposed that defensins are released during a state of active inflammation. Our results show that mice exposed to the high salt diet, previously shown to induce inflammation,Citation48 resulted in an increase in fecal β-defensin secretion () and microbial diversity () without evidence of overt inflammation (). This suggests that altered β-defensin-3 secretion was stimulated by altered gut microbiota composition, resulting in an imbalance between commensal protective and proinflammatory bacteria, induced by high salt. Although T-bet was found to be upregulated, its expression was not accompanied by an upregulation of any other genes related to inflammation, for example, IL-17 that has previously been associated with an increased dietary salt.Citation54,Citation55 Innate immune activation rather than overt inflammation has been implicated in the pathogenesis of at least a subset of IBS patients, and our results are in line with this in patients with dysbiosis.Citation33,Citation56

We recognize that the decreased β-defensin-3 levels following exposure to antimicrobials () and high-fat/high-sugar diet () may appear to be in contradiction to reported single time point elevations in defensin secretion in previous studies.Citation19,Citation33 However, these findings are in line with a report of low β-defensin levels in IBS,Citation35 and these apparent discrepancies may simply reflect the instability of the microbiota in IBS.Citation15–17

Our data indicate that bacterial diversity correlated with β-defensins, suggesting that β-defensin-3 secretion oscillates according to the presence of protective or non-protective (pro-inflammatory) bacteria. A closer inspection of the specific bacterial taxa showed that during antimicrobial exposure bacteria with documented pro-inflammatory effects on the host correlated positively with β-defensin-3 levels. These include bacteria belonging to the genera Eisenbergiella,Citation44 Muribaculum,Citation45 and Prevotellaceae,Citation41,Citation42 (). Additionally, pro-inflammatory Candidatus ArthromitusCitation43 was found to positively correlate with β-defensin-3 levels in both AMC and HFSHD groups, while negatively correlated with β-defensin-3 levels in the HSD group. Its decreased abundance could be a consequence of increased defensin secretion during HSD treatment. Interestingly, Candidatus Arthromitus is a member of Segmented Filamentous Bacteria (SFB) that have been associated with anti-microbial peptide responses and colonization resistance to pathogenic Enterobacteriaceae.Citation43,Citation57,Citation58 Conversely, the proinflammatory bacteria, Eisenbergiella,Citation44 correlated positively with defensin secretion across all groups. This suggests that its low abundance during AMC and HFHSD treatments, and high abundance during HSD treatment, could potentially contribute to the modulation of defensin secretion throughout the treatment periods in all groups. Furthermore, the transcription of inducible defensins such as human β-defensin-2 and -3 has been linked to an increased abundance of species of Prevotellaceae.Citation41,Citation42 Conversely, negative correlations were seen between defensin levels and the abundance of bacteria with both pro- and anti-inflammatory properties like BacteroidesCitation59,Citation60 and AlistipesCitation39,Citation40 (r = −0.5, p = 0.01) (). Thus, our findings suggest that homeostatic regulation of defensin secretion in the gut depends on an intricate balance between both protective and pro-inflammatory bacteria (Supplementary Figure S7).

Our proof-of-principle study has limitations. First, the study was performed in healthy mice. Future studies should concentrate on mice that have an IBS-phenotype with disturbed motility and innate immune activation.Citation19 This is necessary as a bi-directional relationship exists between the microbiota and host physiology.Citation61 Changes in the microbiota alter intestinal permeability,Citation62 motility,Citation63 and visceral sensitivity,Citation38 and conversely, changes, for example, in gut motility, alter microbial composition.Citation64 Dietary change, a common factor in IBS symptom expression, together with changes in gut physiology act together to produce temporal instability of the microbiota in at least a subset of IBS patients.Citation15–17 We propose that studying β-defensin secretion over time in a humanized mouse model of IBSCitation19 in which there is dysbiosis and increased β-defensin-3 would be a logical next step after this proof of concept study and prior to considering clinical studies. A second limitation is the mild nature of the stressor used in this study, as other forms of stress, such as social stress, have been shown to alter microbiota composition and host cytokine levels.Citation65,Citation66 Third, we acknowledge the potential of other epithelial constituents and their secretions that could influence the microbial composition of the gut. For instance, antibiotic treatments have been shown to alter the structure of the mucus layerCitation67 that impacts the diffusion of antimicrobial peptides and immunoglobulins in the small intestine.Citation68 Any alteration in mucus production or composition can contribute to dysbiosis as a result of a breakdown of the gut barrier, hampering intestinal tight junction function and increasing bacterial translocation.Citation69 Epithelial cells produce molecules, such as cytokines, and chemokines, which regulate the gut microbiota and protect against pathogens and thereby influence microbial composition. For example, administering antibiotics to treat DSS-induced colitis in mice worsens the condition by decreasing the microbial ligands that activate Toll-like receptors (TLRs) on epithelial cells, which are crucial for promoting the expression of tissue homeostasis and repair mediators, necessary for maintaining healthy colon function.Citation70 In addition, a high-fat diet has been shown to cause endoplasmic reticulum stress in intestinal epithelial cells, leading to a reduction in claudin-1 expression and mucus barrier function, which can result in increased serum endotoxin levels and dysbiosis.Citation71 The long-term consumption of a high-salt diet has also been associated with altered colonic mucosal immunity, with increased expression of pro-inflammatory genes, and decreased expression of cytokine and chemokine genes in DSS mice.Citation48 These studies suggest that elevated levels of pro-inflammatory cytokines and chemokines can influence β-defensin secretion and thus contribute to dysbiosis.

Our results support the concept that changes in fecal β-defensin-3 act as a marker of altered microbiota composition in mice. Defensin comparisons between baseline and relapse may help identify the presence of dysbiosis in chronic GI conditions like IBS and identify patients who may benefit from microbiota-directed therapies.

Materials and methods

Study design

Four models of experimentally induced dysbiosis were used to determine changes in fecal β-defensin-3 and to characterize the microbiota composition before, during and after each insult, in both males and females. We used (1) an antimicrobial cocktail (AMC) in drinking water, (2) a high-fat/high-sugar diet (HFHSD), (3) a high salt diet (HSD), and (4) mild repeated restraint stress (MRS). In the AMC and dietary studies, we employed a 1-week intervention period which was preceded by a week-long baseline and followed by a recovery period. The same group of mice were followed over the course of 3-week period, and fecal samples were collected every day to measure the fluctuations in each β-defensin-3 (a murine homologue of human β-defensin-2) levels and microbiota. In MRS studies, mice comparisons were made between a control and a stressed group. Fecal samples were collected for consecutively 8 days.

Animals

All experiments were performed using 7- to 10-week-old specific pathogen-free (SPF) C57BL/6 mice except studies using MRS that were performed in NIH Swiss SPF mice. All mice were purchased from Taconic Biosciences and were housed in plastic cages at McMaster University’s central animal facility (CAF) under a 12 h light/dark schedule in a temperature-controlled vivarium with ad libitum access to food and water. Mice received irradiated control diet (1020, Teklad, with 20% of calories from protein, 29% from fat, and 51% from carbohydrates, containing 0.4% Na and 0.7% Cl) and sterile tap water ad libitum upon arrival as well as during baseline and recovery periods. Experiments started 1-week after arrival in the CAF to allow the animals to habituate to the novel environment before initiating the experimental protocols. Cages were changed before and after the initiation of the experiments. The status of the animals was monitored by daily general examination and body weight measurements. Food and water intake was also measured at alternate days. All mice were handled only in a level II biosafety hood to prevent bacterial contamination. Mice were sacrificed using a standardized protocol under Isoflurane (Fresenius Kabi Canada, Toronto, ON) anesthesia. The experiments were approved by the McMaster University animal ethics committee under the Animal Utilization Protocol #18-08-35.

Antimicrobials intervention

The AMC was administered following a previously established protocol.Citation37,Citation38 Briefly, C57BL/6 males (n = 5) and females (n = 5) received a mixture of non-absorbable antimicrobials (neomycin 5 mg/mL, bacitracin 5 mg/mL, and pimaricin 1.25 microg/mL) in their drinking water for 1 week. For the preparation of antimicrobials mixture, neomycin and bacitracin were weighted and stirred in water for 30 min to fully dissolve the powdered antimicrobials before adjusting the pH. At the end, pimaricin was added to the mixture. The antimicrobials were prepared fresh every 48 h.

Dietary interventions

Two different diets were used for our dietary intervention experiments: a high-fat/high-sugar diet (HFHSD) and a high salt diet (HSD). The protocol consisted of two separate groups of mice (n = 5 males and 5 females each) that were fed either HFHSD or HSD for 1 week. The HFHSD was composed of Kcal%: Carb, 35; Fat, 45; Fiber, 0.0 (Prot, 20, # D12451, Research Diets, New Brunswick, NJ), and the HSD was the same as the control diet administered during the baseline period but supplemented with 4% NaCl (TD.130834) ordered from Envigo, Teklad, WI USA. Both groups were administered regular sterile drinking water during the intervention periods.

Mild repeated restraint stress

Both male and female mice were divided into two groups consisting of a control group (n = 4 males and 4 females) and stress group (n = 3 males and 4 females). The mice from the stress group were placed in a plexiglass restrainer for 30 min daily, at a fixed time of the day, for 8 days. The restrained mice were placed in a new cage and were positioned in the restrainers in a way that allowed minimal to no movement but a continued supply of oxygen. Following the stress cycle, mice were transferred back to their home cage and were left undisturbed for the rest of the day. Fecal pellets were collected from both groups every day for the course of the experiment. NIH Swiss mice were used as they have been previously shown to be more sensitive to stress than other mouse strains.Citation72

Assessment of overt inflammation using myeloperoxidase (MPO) activity following HSD

A separate group of mice (n = 5 males and 5 females/timepoint; total mice = 30) followed the same HSD protocol as mentioned above but were sacrificed at the end of every timepoint (baseline, intervention, and recovery). After sacrifice, the entire colon was removed, and tissue samples were snap-frozen in liquid nitrogen for assessment of MPO activity as described previously.Citation73 The MPO activity was expressed in units/milligrams of tissue. Outliers were removed wherever appropriate.

Mouse β-defensin-3 Enzyme Linked Immuno-Sorbent Assay (ELISA)

Mouse β-defensin-3 were measured by ELISA (MyBioSource, MBS034940, San Diego, CA, US) according to manufacturer’s instructions. Briefly, each sample was separately weighted, and PBS was added accordingly (10 µl/mg of sample). Then, samples were mechanically homogenized with 3.0 mm ceramic beads (Sigma, USA) in a bullet homogenizer (Next Advance, NY USA). The samples were homogenized for 10 min at maximum speed, followed by centrifugation at 3000 rpm for 20 min. The supernatants were then collected and loaded on the ELISA plate with blanks and standards. Each sample was run in duplicate. Optical density was read at 450 nm using ELx808 absorbance reader (BioTek, Winooski, VT, USA), and the results were expressed in pg/ml.

DNA isolation and 16S rRNA illumina sequencing

Fecal samples were chosen for Illumina sequencing of the16S rRNA gene, at baseline, day 1 and day 7, during intervention at day 15 and at the end of recovery period at day 21. Fecal samples were homogenized and processed to extract DNA based on previously described protocols.Citation19,Citation48 Polymerase chain reaction (PCR) amplification of the hypervariable 3 (V3) region of the 16S rRNA gene was performed as previously described.Citation74,Citation75 Purified PCR products were sequenced using the Illumina MiSeq platform at the McMaster Genomics facility. Isolated gDNA saved from the Illumina sequencing protocol was used to quantify bacterial load using RT-PCR. Briefly, gDNA was collected using one fecal pellet per time point. Amplifications were performed on Step one plus real-time PCR system (BioRad, ON, CA) (95°C for 10 min, followed by 40 cycles of 95°C for 15 s, 55°C for 40 s and 62°C for 60 s) using SYBR Green-based PCR master mix (Applied Biosystems) and the following primers 926 F (5’ AAA CTC AAA KGA ATT GAC GG) at E. coli position 908–926 and 1062 F (5’ CTC ACR RCA CGA GCT GAC) at E. coli position 1081–1064 targeting regions V6-V8, as described previously.Citation76 Samples were run in duplicates, and data were normalized with reference to baseline (day 1 and day 7) timepoint measurements and analyzed using the two-ΔCt method. A melting analysis was also carried out following the last cycle of each amplification to confirm the amplification specificity. For each experimental group (AMC, HFHSD, HSD), a standard curve with efficiency between 91% and 100% was run together with the samples, as stated earlier.Citation19

16S rRNA processing and analysis/16S Ribosomal RNA gene analysis

All sequences obtained following sequencing were processed using Cutadapt to trim and filter adapter sequences and PCR primers from the raw reads with a minimum quality score of 30 and minimum read length of 100 base pairs, as previously described.Citation19 Further, denoising Algorithm 2 (DADA 2) package in R was used to align the paired-end sequences and perform all the subsequent quality filtering and removing chimeras.Citation77 Taxonomy was assigned using the Silva reference database.Citation78 ASVs not assigned to bacterial genera or assigned to either mitochondria or chloroplast were removed from analysis. Any ASVs identified as chimeric by de novo detection in DADA2 R package were removed. Mouse samples had an average of 69,846.46 reads per sample after filtering, ranging from 11,055 to 171,477 reads per sample. The analysis was performed by merging all Illumina runs, running all analyses together and then sub-setting the samples according to each experimental group used in the study when required. Phyloseq and vegan packages for R, version 4.1.1 (R foundation for Statistical Computing) were used for statistical analyses. Following the filtering of non-bacterial taxa from samples, principal coordinate analysis (PCoA) was performed using the Bray-Curtis and Aitchison Distance matrices to evaluate beta-diversity. Statistical differences in beta-diversity between the experimental groups were analyzed using the ADONIS2 function in the vegan package. Reads from baseline and days 1 and 7 were combined and considered as a single baseline timepoint. To evaluate differences in alpha diversity, Shannon diversity was employed in the absence of rarefaction. Differences in alpha diversity were assessed using one-way ANOVA with FDR correction for multiple comparisons using Dunn’s test. The identification of any differentially abundant bacterial taxa associated with baseline, intervention and recovery periods was conducted using the ANCOM-BC package,Citation79 after filtering the dataset to remove any ASVs with a frequency of less than 4 in at least 10% of the samples and converting the absolute abundance to relative abundance. The samples were then aggregated at the genus level, merging bacteria belonging to the same strain into a single entity. Statistical significance on differentially abundant taxa between groups was corrected for multiple comparisons using false discovery rate (FDR). False discovery rate with a Q value of 0.05 was used for the multiple testing of bacterial taxa. Color intensity on heatmaps was generated by using z-scores calculated relatively within each data set (bacteria). PCoA plots, heatmaps and diversity box plots were plotted using phyloseq, vegan and ggplot packages for R or using GraphPad Prism version 9.

Gene expression analysis

RNeasy Mini Kit (Qiagen, Germany) was used to extract and purify total RNA from whole colonic tissue samples according to manufacturer’s instructions. Additional steps were performed during purification to remove DNA traces from the samples using the RNease-Free DNase Set (Qiagen). A custom Code Set panel that included 68 genes related to immune, gut barrier, and neurobiology functions was run according to the manufacturer’s instructions (NanoString Technologies Inc., Seattle, WA, USA), and data were analyzed with nSolver 4.0 software (Nanostring Technologies Inc.). The log2 ratios built from the data obtained were then plotted as volcano plots and represented as heatmaps.

Data analysis

Statistical analysis was carried out using GraphPad Prism 9 (GraphPad Software, La Jolla, CA, USA), and R (version 4.1.1). Statistical comparisons were performed using one-way ANOVA, two-way ANOVA, Kruskal−Wallis, Mann−Whitney or unpaired t-test, as appropriate. Dunn’s test or Benjamini, Krieger and Yekutieli were used, when performing post hoc tests for multiple comparisons. A non-parametric Spearman rank test followed by FDR corrections was used when performing correlations. Two-tailed statistical tests were used. A p-value of less than 0.05 was considered to be statistically significant.

Author contribution

ZS performed all the experiments and data analyses. SMC designed and supervised the project. PB provided scientific input and assistance with writing. GDP critically reviewed the manuscript. MS provided assistance with microbiota analyses.

Supplemental material

Supplemental Material

Download Zip (1.1 MB)

Acknowledgments

The authors thank Central Animal Facility at McMaster (CAF) staff for their support with murine experiments. The authors also thank Vidhya Mohan (PhD student under PB) for her assistance with microbiota analysis.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Data availability statement

The raw 16S rRNA sequences and the gene expression data have been deposited at NCBI under the umbrella BioProject PRJNA935320. Individual public projects under this BioProject are SRA BioProject (PRJNA932045) and Geo BioProject (PRJNA934868). Supplemental data that supports the findings of this study can be accessed online at https://doi.org/10.6084/m9.figshare.23116457.v1.

Supplementary material

Supplemental data for this article can be accessed online at https://doi.org/10.1080/19490976.2023.2233679

Additional information

Funding

This study was supported by a Canadian Institutes of Health Foundation grant # 20001944 to SMC and PB.

References

  • Dethlefsen L, Huse S, Sogin ML, Relman DA, Eisen JA. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol. 2008;6(11):e280. doi:10.1371/journal.pbio.0060280.
  • Dethlefsen L, Relman DA. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc Natl Acad Sci USA. 2011;Suppl 108(Suppl supplement_1):4554–19. doi:10.1073/pnas.1000087107.
  • Campana AM, Laue HE, Shen Y, Shrubsole MJ, Baccarelli AA. Assessing the role of the gut microbiome at the interface between environmental chemical exposures and human health: Current knowledge and challenges. Environ Pollut. 2022;120380. doi:10.1016/j.envpol.2022.120380.
  • Mendall MA, Kumar D. Antibiotic use, childhood affluence and irritable bowel syndrome (IBS). Eur J Gastroenterol Hepatol. 1998;10(1):59–62. doi:10.1097/00042737-199801000-00011.
  • Maxwell PR, Rink E, Kumar D, Mendall MA. Antibiotics increase functional abdominal symptoms. Am J Gastroenterol. 2002;97(1):104–108. doi:10.1111/j.1572-0241.2002.05428.x.
  • Gibson PR, Barrett JS, Muir JG. Functional bowel symptoms and diet. Intern Med J. 2013;43(10):1067–1074. doi:10.1111/imj.12266.
  • Simrén M, Månsson A, Langkilde AM, Svedlund J, Abrahamsson H, Bengtsson U, Björnsson ES. Food-related gastrointestinal symptoms in the irritable bowel syndrome. Digestion. 2001;63(2):108–115. doi:10.1159/000051878.
  • Na W, Lee Y, Kim H, Kim YS, Sohn C. High-fat foods and FODMAPs containing gluten foods primarily contribute to symptoms of irritable bowel syndrome in Korean adults. Nutrients. 2021;13(4). doi:10.3390/nu13041308.
  • Zoetendal EG, Akkermans AD, De Vos WM. Temperature gradient gel electrophoresis analysis of 16S rRNA from human fecal samples reveals stable and host-specific communities of active bacteria. Appl Environ Microbiol. 1998;64(10):3854–3859. doi:10.1128/aem.64.10.3854-3859.1998.
  • Caporaso JG, Lauber CL, Costello EK, Berg-Lyons D, Gonzalez A, Stombaugh J, Knights D, Gajer P, Ravel J, Fierer N, et al. Moving pictures of the human microbiome. Genome Biol. 2011;12(5):R50. doi:10.1186/gb-2011-12-5-r50.
  • Hisada T, Endoh K, Kuriki K. Inter- and intra-individual variations in seasonal and daily stabilities of the human gut microbiota in Japanese. Arch Microbiol. 2015;197(7):919–934. doi:10.1007/s00203-015-1125-0.
  • Schloissnig S, Arumugam M, Sunagawa S, Mitreva M, Tap J, Zhu A, Waller A, Mende DR, Kultima JR, Martin J, et al. Genomic variation landscape of the human gut microbiome. Nature. 2013;493(7430):45–50. doi:10.1038/nature11711.
  • Faith JJ, Guruge JL, Charbonneau M, Subramanian S, Seedorf H, Goodman AL, Clemente JC, Knight R, Heath AC, Leibel RL, et al. The long-term stability of the human gut microbiota. Science. 2013;341(6141):1237439. doi:10.1126/science.1237439.
  • Mehta RS, Abu-Ali GS, Drew DA, Lloyd-Price J, Subramanian A, Lochhead P, Joshi AD, Ivey KL, Khalili H, Brown GT, et al. Stability of the human faecal microbiome in a cohort of adult men. Nature Microbiology. 2018;3(3):347–355. doi:10.1038/s41564-017-0096-0.
  • Mättö J, Maunuksela L, Kajander K, Palva A, Korpela R, Kassinen A, Saarela M. Composition and temporal stability of gastrointestinal microbiota in irritable bowel syndrome–a longitudinal study in IBS and control subjects. FEMS Immunol Med Microbiol. 2005;43(2):213–222. doi:10.1016/j.femsim.2004.08.009.
  • Maukonen J, Satokari R, Mättö J, Söderlund H, Mattila-Sandholm T, Saarela M. Prevalence and temporal stability of selected clostridial groups in irritable bowel syndrome in relation to predominant faecal bacteria. J Med Microbiol. 2006;55(Pt 5):625–633. doi:10.1099/jmm.0.46134-0.
  • Durbán A, Abellán JJ, Jiménez-Hernández N, Artacho A, Garrigues V, Ortiz V, Ponce J, Latorre A, Moya A. Instability of the faecal microbiota in diarrhoea-predominant irritable bowel syndrome. FEMS Microbiol Ecol. 2013;86(3):581–589. doi:10.1111/1574-6941.12184.
  • Myneedu K, Deoker A, Schmulson MJ, Bashashati M. Fecal microbiota transplantation in irritable bowel syndrome: A systematic review and meta-analysis. United European Gastroenterol J. 2019;7(8):1033–1041. doi:10.1177/2050640619866990.
  • De Palma G, Lynch MD, Lu J, Dang VT, Deng Y, Jury J, Umeh G, Miranda PM, Pigrau Pastor M, Sidani S, et al. Transplantation of fecal microbiota from patients with irritable bowel syndrome alters gut function and behavior in recipient mice. Sci Transl Med. 2017;9(379). doi:10.1126/scitranslmed.aaf6397.
  • Fusco A, Savio V, Cammarota M, Alfano A, Schiraldi C, Donnarumma G. Beta-defensin-2 and beta-defensin-3 reduce intestinal damage caused by salmonella typhimurium modulating the expression of cytokines and enhancing the probiotic activity of enterococcus faecium. J Immunol Res. 2017;2017(6976935):1–9. doi:10.1155/2017/6976935.
  • Brown EM, Sadarangani M, Finlay BB. The role of the immune system in governing host-microbe interactions in the intestine. Nat Immunol. 2013;14(7):660–667. doi:10.1038/ni.2611.
  • Dhople V, Krukemeyer A, Ramamoorthy A. The human beta-defensin-3, an antibacterial peptide with multiple biological functions. Biochim Biophys Acta. 2006;1758(9):1499–1512. doi:10.1016/j.bbamem.2006.07.007.
  • Dhawan S, De Palma G, Willemze RA, Hilbers FW, Verseijden C, Luyer MD, Nuding S, Wehkamp J, Souwer Y, de Jong EC, et al. Acetylcholine-producing T cells in the intestine regulate antimicrobial peptide expression and microbial diversity. Am J Physiol Gastrointest Liver Physiol. 2016;311(5):G920–g933. doi:10.1152/ajpgi.00114.2016.
  • Meade KG, O’Farrelly C. β-defensins: Farming the microbiome for homeostasis and health. Front Immunol. 2018;9:3072. doi:10.3389/fimmu.2018.03072.
  • Kim JM. Antimicrobial proteins in intestine and inflammatory bowel diseases. Intest Res. 2014;12(1):20–33. doi:10.5217/ir.2014.12.1.20.
  • O’Neil DA, Porter EM, Elewaut D, Anderson GM, Eckmann L, Ganz T, Kagnoff MF. Expression and regulation of the human beta-defensins Hbd-1 and Hbd-2 in intestinal epithelium. J Immunol. 1999;163(12):6718–6724. doi:10.4049/jimmunol.163.12.6718.
  • Wehkamp J, Fellermann K, Herrlinger KR, Baxmann S, Schmidt K, Schwind B, Duchrow M, Wohlschläger C, Feller AC, Stange EF. Human beta-defensin 2 but not beta-defensin 1 is expressed preferentially in colonic mucosa of inflammatory bowel disease. Eur J Gastroenterol Hepatol. 2002;14(7):745–752. doi:10.1097/00042737-200207000-00006.
  • Fusco A, Savio V, Donniacuo M, Perfetto B, Donnarumma G. Antimicrobial peptides human beta-defensin-2 and -3 protect the gut during Candida albicans infections enhancing the intestinal barrier integrity: In Vitro study. Front Cell Infect Microbiol. 2021;11(666900). doi:10.3389/fcimb.2021.666900.
  • Ayala-Sumuano JT, Téllez-López VM, Domínguez-Robles Mdel C, Shibayama-Salas M, Meza I, Singer SM. Toll-like receptor signaling activation by Entamoeba histolytica induces beta defensin 2 in human colonic epithelial cells: its possible role as an element of the innate immune response. PLoS Negl Trop Dis. 2013;7(2):e2083. doi:10.1371/journal.pntd.0002083.
  • Salzman NH, Hung K, Haribhai D, Chu H, Karlsson-Sjöberg J, Amir E, Teggatz P, Barman M, Hayward M, Eastwood D, et al. Enteric defensins are essential regulators of intestinal microbial ecology. Nat Immunol. 2010;11(1):76–83. doi:10.1038/ni.1825.
  • Masuda K, Sakai N, Nakamura K, Yoshioka S, Ayabe T. Bactericidal activity of mouse α-defensin cryptdin-4 predominantly affects noncommensal bacteria. J Innate Immun. 2011;3(3):315–326. doi:10.1159/000322037.
  • Riba A, Olier M, Lacroix-Lamandé S, Lencina C, Bacquié V, Harkat C, Gillet M, Baron M, Sommer C, Mallet V, et al. Paneth cell defects induce microbiota dysbiosis in mice and promote visceral hypersensitivity. Gastroenterology. 2017;153(6):1594–1606.e2. doi:10.1053/j.gastro.2017.08.044.
  • Langhorst J, Junge A, Rueffer A, Wehkamp J, Foell D, Michalsen A, Musial F, Dobos GJ. Elevated human beta-defensin-2 levels indicate an activation of the innate immune system in patients with irritable bowel syndrome. Am J Gastroenterol. 2009;104(2):404–410. doi:10.1038/ajg.2008.86.
  • Shulman RJ, Devaraj S, Heitkemper M. Activation of the innate immune system in children with irritable bowel syndrome evidenced by increased fecal human β-defensin-2. Clin Gastroenterol Hepatol. 2021;19(10):2121–2127. doi:10.1016/j.cgh.2020.09.034.
  • Mujagic Z, Tigchelaar EF, Zhernakova A, Ludwig T, Ramiro-Garcia J, Baranska A, Swertz MA, Masclee AA, Wijmenga C, van Schooten FJ, et al. A novel biomarker panel for irritable bowel syndrome and the application in the general population. Sci Rep. 2016;6:26420. doi:10.1038/srep26420.
  • Gacesa R, Vich Vila A, Collij V, Mujagic Z, Kurilshikov A, Voskuil MD, Festen EAM, Wijmenga C, Jonkers D, Dijkstra G, et al. A combination of fecal calprotectin and human beta-defensin 2 facilitates diagnosis and monitoring of inflammatory bowel disease. Gut Microbes. 2021;13(1):1943288. doi:10.1080/19490976.2021.1943288.
  • Bercik P, Denou E, Collins J, Jackson W, Lu J, Jury J, Deng Y, Blennerhassett P, Macri J, McCoy KD, et al. The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology. 2011;141(2):599-609, 609.e1–3. doi:10.1053/j.gastro.2011.04.052.
  • Verdú EF, Bercik P, Verma-Gandhu M, Huang XX, Blennerhassett P, Jackson W, Mao Y, Wang L, Rochat F, Collins SM. Specific probiotic therapy attenuates antibiotic induced visceral hypersensitivity in mice. Gut. 2006;55(2):182–190. doi:10.1136/gut.2005.066100.
  • Parker BJ, Wearsch PA, Veloo ACM, Rodriguez-Palacios A. The genus alistipes: Gut bacteria with emerging implications to inflammation, cancer, and mental health. Front Immunol. 2020;11:906. doi:10.3389/fimmu.2020.00906.
  • El-Salhy M, Mazzawi T, Hausken T, Hatlebakk JG. Irritable bowel syndrome patients who are not likely to respond to fecal microbiota transplantation. Neurogastroenterol Motil. 2022;34(9):e14353. doi:10.1111/nmo.14353.
  • Levy M, Thaiss CA, Zeevi D, Dohnalová L, Zilberman-Schapira G, Mahdi JA, David E, Savidor A, Korem T, Herzig Y, et al. Microbiota-modulated metabolites shape the intestinal microenvironment by regulating NLRP6 inflammasome signaling. Cell. 2015;163(6):1428–1443. doi:10.1016/j.cell.2015.10.048.
  • Scher JU, Sczesnak A, Longman RS, Segata N, Ubeda C, Bielski C, Rostron T, Cerundolo V, Pamer EG, Abramson SB, et al. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife. 2013;2:e01202. doi:10.7554/eLife.01202.
  • Santiago A, Sanchez E, Clark A, Pozuelo M, Calvo M, Yañez F, Sarrabayrouse G, Perea L, Vidal S, Gallardo A, et al. Sequential changes in the mesenteric lymph node microbiome and immune response during Cirrhosis induction in Rats. mSystems. 2019;4(1). doi:10.1128/mSystems.00278-18
  • Li HY, Zhang HL, Zhao FJ, Wang SQ, Wang ZX, Wei ZY. Modulation of gut microbiota, short-chain fatty acid production, and inflammatory cytokine expression in the cecum of porcine deltacoronavirus-infected chicks. Front Microbiol. 2020;11:897. doi:10.3389/fmicb.2020.00897.
  • Obermüller B, Singer G, Kienesberger B, Klymiuk I, Sperl D, Stadlbauer V, Horvath A, Miekisch W, Gierschner P, Grabherr R, et al. The Effects of Prebiotic Supplementation with OMNi-LOGiC® FIBRE on Fecal Microbiome, Fecal Volatile Organic Compounds, and Gut Permeability in Murine Neuroblastoma-Induced Tumor-Associated Cachexia. Nutrients. 2020;12(7):2029. doi:10.3390/nu12072029.
  • Ma L, Shen Q, Lyu W, Lv L, Wang W, Yu M, Yang H, Tao S, Xiao Y, Claesen J. Clostridium butyricum and its derived extracellular vesicles modulate gut homeostasis and ameliorate acute experimental colitis. Microbiol Spectr. 2022;10(4):e0136822. doi:10.1128/spectrum.01368-22.
  • Aghamohammad S, Sepehr A, Miri ST, Najafi S, Pourshafie MR, Rohani M. Anti-inflammatory and immunomodulatory effects of Lactobacillus spp. as a preservative and therapeutic agent for IBD control. Immun Inflamm Dis. 2022;10(6):e635. doi:10.1002/iid3.635.
  • Miranda PM, De Palma G, Serkis V, Lu J, Louis-Auguste MP, McCarville JL, Verdu EF, Collins SM, Bercik P. High salt diet exacerbates colitis in mice by decreasing Lactobacillus levels and butyrate production. Microbiome. 2018;6(1):57. doi:10.1186/s40168-018-0433-4.
  • Garrido-Mesa N, Schroeder JH, Stolarczyk E, Gallagher AL, Lo JW, Bailey C, Campbell L, Sexl V, MacDonald TT, Howard JK, et al. T-bet controls intestinal mucosa immune responses via repression of type 2 innate lymphoid cell function. Mucosal Immunol. 2019;12(1):51–63. doi:10.1038/s41385-018-0092-6.
  • Jeffery IB, O’Toole PW, Öhman L, Claesson MJ, Deane J, Quigley EM, Simrén M. An irritable bowel syndrome subtype defined by species-specific alterations in faecal microbiota. Gut. 2012;61(7):997–1006. doi:10.1136/gutjnl-2011-301501.
  • Collins SM. A role for the gut microbiota in IBS. Nat Rev Gastroenterol Hepatol. 2014;11(8):497–505. doi:10.1038/nrgastro.2014.40.
  • Wehkamp J, Harder J, Weichenthal M, Mueller O, Herrlinger KR, Fellermann K, Schroeder JM, Stange EF. Inducible and constitutive beta-defensins are differentially expressed in Crohn’s disease and ulcerative colitis. Inflamm Bowel Dis. 2003;9(4):215–223. doi:10.1097/00054725-200307000-00001.
  • Zilbauer M, Jenke A, Wenzel G, Postberg J, Heusch A, Phillips AD, Noble-Jamieson G, Torrente F, Salvestrini C, Heuschkel R, et al. Expression of human beta-defensins in children with chronic inflammatory bowel disease. PLoS One. 2010;5(10):e15389. doi:10.1371/journal.pone.0015389.
  • Kleinewietfeld M, Manzel A, Titze J, Kvakan H, Yosef N, Linker RA, Muller DN, Hafler DA. Sodium chloride drives autoimmune disease by the induction of pathogenic TH17 cells. Nature. 2013;496(7446):518–522. doi:10.1038/nature11868.
  • Wu C, Yosef N, Thalhamer T, Zhu C, Xiao S, Kishi Y, Regev A, Kuchroo VK. Induction of pathogenic TH17 cells by inducible salt-sensing kinase SGK1. Nature. 2013;496(7446):513–517. doi:10.1038/nature11984.
  • Downs IA, Aroniadis OC, Kelly L, Brandt LJ. Postinfection irritable bowel syndrome: The links between gastroenteritis, inflammation, the microbiome, and functional disease. J Clin Gastroenterol. 2017;51(10):869–877. doi:10.1097/mcg.0000000000000924.
  • Zheng Y, Valdez PA, Danilenko DM, Hu Y, Sa SM, Gong Q, Abbas AR, Modrusan Z, Ghilardi N, de Sauvage FJ, et al. Interleukin-22 mediates early host defense against attaching and effacing bacterial pathogens. Nat Med. 2008;14(3):282–289. doi:10.1038/nm1720.
  • Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009;139(3):485–498. doi:10.1016/j.cell.2009.09.033.
  • Yamanouchi Y, Chudan S, Ishibashi R, Ohue-Kitano R, Nishikawa M, Tabuchi Y, Kimura I, Nagai Y, Ikushiro S, Furusawa Y. The impact of low-viscosity soluble dietary fibers on intestinal microenvironment and experimental colitis: a possible preventive application of alpha-cyclodextrin in intestinal inflammation. Mol Nutr Food Res. 2022;66(22):e2200063. doi:10.1002/mnfr.202200063.
  • Hiippala K, Kainulainen V, Suutarinen M, Heini T, Bowers JR, Jasso-Selles D, Lemmer D, Valentine M, Barnes R, Engelthaler DM, et al. Isolation of anti-inflammatory and epithelium reinforcing bacteroides and parabacteroides spp. from a healthy fecal donor. Nutrients. 2020;12(4). doi:10.3390/nu12040935.
  • Agirman G, Hsiao EY. SnapShot: The microbiota-gut-brain axis. Cell. 2021;184(9):2524–2524.e1. doi:10.1016/j.cell.2021.03.022.
  • Willing BP, Russell SL, Finlay BB. Shifting the balance: antibiotic effects on host-microbiota mutualism. Nat Rev Microbiol. 2011;9(4):233–243. doi:10.1038/nrmicro2536.
  • Anitha M, Vijay-Kumar M, Sitaraman SV, Gewirtz AT, Srinivasan S. Gut microbial products regulate murine gastrointestinal motility via Toll-like receptor 4 signaling. Gastroenterology. 2012;143(4):1006–16.e4. doi:10.1053/j.gastro.2012.06.034.
  • Kashyap PC, Marcobal A, Ursell LK, Larauche M, Duboc H, Earle KA, Sonnenburg ED, Ferreyra JA, Higginbottom SK, Million M, et al. Complex interactions among diet, gastrointestinal transit, and gut microbiota in humanized mice. Gastroenterology. 2013;144(5):967–977. doi:10.1053/j.gastro.2013.01.047.
  • Bailey MT, Dowd SE, Galley JD, Hufnagle AR, Allen RG, Lyte M. Exposure to a social stressor alters the structure of the intestinal microbiota: implications for stressor-induced immunomodulation. Brain Behav Immun. 2011;25(3):397–407. doi:10.1016/j.bbi.2010.10.023.
  • Maltz RM, Keirsey J, Kim SC, Mackos AR, Gharaibeh RZ, Moore CC, Xu J, Somogyi A, Bailey MT. Social stress affects colonic inflammation, the gut microbiome, and short-chain fatty acid levels and receptors. J Pediatr Gastroenterol Nutr. 2019;68(4):533–540. doi:10.1097/mpg.0000000000002226.
  • Chen CY, Hsu KC, Yeh HY, Ho HC. Visualizing the effects of antibiotics on the mouse colonic mucus layer. Ci Ji Yi Xue Za Zhi. 2020;32(2):145–153. doi:10.4103/tcmj.tcmj_70_19.
  • Johansson ME, Jakobsson HE, Holmén-Larsson J, Schütte A, Ermund A, Rodríguez-Piñeiro AM, Arike L, Wising C, Svensson F, Bäckhed F, et al. Normalization of host intestinal mucus layers requires long-term microbial colonization. Cell Host & Microbe. 2015;18(5):582–592. doi:10.1016/j.chom.2015.10.007.
  • Feng Y, Huang Y, Wang Y, Wang P, Song H, Wang F, Weber CR. Antibiotics induced intestinal tight junction barrier dysfunction is associated with microbiota dysbiosis, activated NLRP3 inflammasome and autophagy. PLoS One. 2019;14(6):e0218384. doi:10.1371/journal.pone.0218384.
  • Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004;118(2):229–241. doi:10.1016/j.cell.2004.07.002.
  • Gulhane M, Murray L, Lourie R, Tong H, Sheng YH, Wang R, Kang A, Schreiber V, Wong KY, Magor G, et al. High fat diets induce colonic epithelial cell stress and inflammation that is reversed by IL-22. Sci Rep. 2016;6:28990. doi:10.1038/srep28990.
  • Macedo GC, Morita GM, Domingues LP, Favoretto CA, Suchecki D, Quadros IMH. Consequences of continuous social defeat stress on anxiety- and depressive-like behaviors and ethanol reward in mice. Horm Behav. 2018;97:154–161. doi:10.1016/j.yhbeh.2017.10.007.
  • Bercík P, De Giorgio R, Blennerhassett P, Verdú EF, Barbara G, Collins SM. Immune-mediated neural dysfunction in a murine model of chronic Helicobacter pylori infection. Gastroenterology. 2002;123(4):1205–1215. doi:10.1053/gast.2002.36024.
  • Bartram AK, Lynch MD, Stearns JC, Moreno-Hagelsieb G, Neufeld JD. Generation of multimillion-sequence 16S rRNA gene libraries from complex microbial communities by assembling paired-end illumina reads. Appl Environ Microbiol. 2011;77(11):3846–3852. doi:10.1128/aem.02772-10.
  • Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJ, Holmes SP. DADA2: High-resolution sample inference from Illumina amplicon data. Nat Methods. 2016;13(7):581–583. doi:10.1038/nmeth.3869.
  • Ghyselinck J, Pfeiffer S, Heylen K, Sessitsch A, De Vos P, Ravel J. The effect of primer choice and short read sequences on the outcome of 16S rRNA gene based diversity studies. PLoS One. 2013;8(8):e71360. doi:10.1371/journal.pone.0071360.
  • Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJ, Holmes SP. DADA2: High-resolution sample inference from Illumina amplicon data. Nat Methods. 2016;13 (7):581–583. doi:10.1038/nmeth.3869.
  • Quast C, Pruesse E, Yilmaz P, Gerken J, Schweer T, Yarza P, Peplies J, Glöckner,FO. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 2013;41(Database issue):D590–6. doi:10.1093/nar/gks1219.
  • Lin H, Peddada SD. Analysis of compositions of microbiomes with bias correction. Nat Commun. 2020;11(1):3514. doi:10.1038/s41467-020-17041-7.