Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2023 Mar 21;4(3):100979.
doi: 10.1016/j.xcrm.2023.100979.

Gut microbe-derived milnacipran enhances tolerance to gut ischemia/reperfusion injury

Affiliations

Gut microbe-derived milnacipran enhances tolerance to gut ischemia/reperfusion injury

Fan Deng et al. Cell Rep Med. .

Abstract

There are significant differences in the susceptibility of populations to intestinal ischemia/reperfusion (I/R), but the underlying mechanisms remain elusive. Here, we show that mice exhibit significant differences in susceptibility to I/R-induced enterogenic sepsis. Notably, the milnacipran (MC) content in the enterogenic-sepsis-tolerant mice is significantly higher. We also reveal that the pre-operative fecal MC content in cardiopulmonary bypass patients, including those with intestinal I/R injury, is associated with susceptibility to post-operative gastrointestinal injury. We reveal that MC attenuates mouse I/R injury in wild-type mice but not in intestinal epithelial aryl hydrocarbon receptor (AHR) gene conditional knockout mice (AHRflox/flox) or IL-22 gene deletion mice (IL-22-/-). Collectively, our results suggest that gut microbiota affects susceptibility to I/R-induced enterogenic sepsis and that gut microbiota-derived MC plays a pivotal role in tolerance to intestinal I/R in an AHR/ILC3/IL-22 signaling-dependent manner, revealing the pathological mechanism, potential prevention and treatment drugs, and treatment strategies for intestinal I/R.

Keywords: IL-22; aryl hydrocarbon receptor; enterogenic sepsis; intestinal flora; milnacipran.

PubMed Disclaimer

Conflict of interest statement

Declaration of interests The authors declare that they have no conflict of interests.

Figures

None
Graphical abstract
Figure 1
Figure 1
Mice have significant differences in susceptibility to intestinal I/R-induced enterogenic sepsis (A) Schematic diagram of differences in susceptibility to enterogenic sepsis in mice. (B) Mouse survival curve (n = 20; chi-square test). (C and D) Hematoxylin and eosin (HE) staining (C) and the pathological damage scores (D) of intestinal tissue sections. Scale bar, 100 μm (n = 8). (E–G) The fluorescein isothiocyanate (FITC)-dextran levels (E) and 16S/18S levels in blood cells (F) and endotoxin levels in plasma (G) reflect the permeability of the intestinal barrier and bacterial translocation (n = 8). (H–J) Mouse lung tissue HE staining (H); the pathological damage scores (I) and wet/dry weight ratios (J). Scale bar, 100 μm (n = 8). (K–N) Mouse liver tissue HE staining (K); the pathological damage scores (L) and plasma ALT (M) and AST levels (N). Scale bar, 100 μm (n = 8). (O–Q) Mouse kidney tissue HE staining (O); the pathological damage scores (P) and plasma BUN levels (Q). Scale bar, 100 μm (n = 8). The results are expressed as the median and quartile. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001 by were determined by the Mann-Whitney test.
Figure 2
Figure 2
Differences in the gut microbiota and metabolites in mice with different susceptibilities to enterogenic sepsis (A) Alpha diversity analysis (Chao1 index, n = 6; Wilcoxon test). (B) Principal coordinate analysis (PCoA) of 16S rRNA gene sequencing data (n = 6; Adonis analysis and Anosim analysis). (C) The composition of the gut bacterial abundance at the phylum level (n = 6). (D) Linear discriminant analysis (LDA) effect size (LEfSe) of different species between groups (n = 6). (E) PCoA of metabolomics (n = 6; Adonis analysis and Anosim analysis). (F) Volcano plot of differential metabolites between groups (n = 6). (G) The differential metabolite Kyoto Encyclopedia of Genes and Genomes (KEGG) bubble plot between groups (n = 6). (H) Chemical structure of MC (n = 6). (I) Liquid chromatography-tandem mass spectrometry (LC–MS/MS)-targeted detection of the MC content in feces of WT mice or germ-free (GF) mice (n = 6). (J) LC-MS/MS-targeted detection of the MC content in the feces. The results are expressed as the median and quartile. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001 by were determined by the Mann-Whitney test in (A), (I), (J), and adonis analysis and anosim analyses in (B) and (E).
Figure 3
Figure 3
Gut microbiota affects susceptibility to enterogenic sepsis induced by intestinal I/R (A) Feces from the ESTM or the ESSM were transplanted into pseudosterile mice to establish a model of enterogenic sepsis. (B) The relative abundance of Bacteroidetes and Verrucomicrobiota in the feces. (C and D) Hematoxylin-eosin (HE) staining (C) and pathological damage scores (D) of intestinal tissue sections. Scale bar, 100 μm. (E–G) The fluorescein isothiocyanate (FITC)-dextran levels (E) and 16S/18S levels in blood cells (F) and endotoxin levels in plasma (G) reflect the permeability of the intestinal barrier, bacterial translocation, and infection. (H) The pre-operative stools of patients undergoing CPB from the AGI group or the NAGI group were transplanted into pseudosterile mice to establish a model of enterogenic sepsis. (I) The MC content in the feces 7 days after transplantation. (J and K) HE staining (J) and pathological damage scores (K) of intestinal tissue sections. Scale bar, 100 μm. (L–N) The FITC-dextran levels (L) and 16S/18S levels in blood cells (M) and endotoxin levels in plasma (N) reflect the permeability of the intestinal barrier, bacterial translocation. The results are expressed as the median and quartile, n = 8 for each group. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001 were determined by the Mann-Whitney test.
Figure 4
Figure 4
MC ameliorates intestinal I/R injury in mice by activating the intestinal epithelial AHR (A and B) Hematoxylin-eosin (HE) staining (A) and pathological damage scores (B) of intestinal tissue sections. Scale bar, 100 μm (n = 8). (C–E) The fluorescein isothiocyanate (FITC)-dextran levels (C), and 16S/18S levels in blood cells (D) endotoxin levels in plasma (E) reflect the permeability of the intestinal barrier and bacterial translocation (n = 8). (F and G) Immunofluorescence (F) and relative quantitative analysis (G) of the cell proliferation marker Ki67 in the intestine. Scale bar, 100 μm (n = 5). (H) Relative mRNA level of the intestinal stem cell self-renewal marker Lgr5 in the intestine (n = 8). (I) HE staining of intestinal organoids. Scale bar, 20 μm (n = 6). (J) Intestinal organoid viability (n = 6). (K) LDH levels in the medium (n = 6). (L–M) Immunofluorescence (L) and relative quantitative analysis (M) of Ki67 in intestinal organoids. Scale bar, 20 μm (n = 5). (N) Relative mRNA level of Ki67 in intestinal organoids (n = 6). (O) Relative mRNA level of Lgr5 in intestinal organoids (n = 6). The results are expressed as the median and quartile. ∗, #, and & p < 0.05; ∗∗, ##, and && p < 0.01; ∗∗∗, ###, and &&& p < 0.001 were determined by two-way ANOVA and Tukey’s post hoc test. ∗p < 0.05 compared with WT mouse Sham group; #p < 0.05 compared with WT mouse I/R group; &p < 0.05 compared with WT mouse I/R + MC group. I/R + MC group: WT mice or AHRflox/flox mice were injected intraperitoneally with 10 mg/kg MC 1 h before intestinal I/R; H/R + MC group: 10 μmol/L MC was added to intestinal organoids 1 h before H/R.
Figure 5
Figure 5
MC promotes increased NCR+ILC3/ILC3 ratio and IL-22 release by activating intestinal epithelial AHR (A and B) Flow cytometry to detect the ratio of NCR+ILC3s/ILC3s (n = 3). (C and D) Flow cytometry to detect the ratio of IL-22+NCR+ILC3s/NCR+ILC3s (n = 3). (E) The mRNA levels of IL-22 in intestinal tissue (n = 8). (F and G) Immunohistochemical results (F) and relative quantitative analysis (G) of IL-22 in intestinal tissue (n = 5). (H) IL-22 levels in intestinal organoid culture medium (n = 6). (I) The 7-day survival curve of mice (n = 20; chi-square test). The results are expressed as the median and quartile. ∗, #, and & p < 0.05; ∗∗, ##, and && p < 0.01; ∗∗∗, ###, and &&& p < 0.001 were determined by two-way ANOVA and Tukey’s post hoc test. ∗p < 0.05 compared with WT mouse Sham group; #p < 0.05 compared with WT mouse I/R group; &p < 0.05 compared with WT mouse I/R + MC group.
Figure 6
Figure 6
AHR activator Ficz reduces intestinal I/R injury in mice by releasing IL-22 (A and B) Hematoxylin-eosin (HE) staining (A) and the pathological damage score (B) of intestinal tissue sections. Scale bar, 100 μm (n = 8). (C–E) The fluorescein isothiocyanate (FITC)-dextran levels (C) and 16S/18S levels in blood cells (D) and endotoxin levels in plasma (E) reflect the permeability of the intestinal barrier and bacterial translocation (n = 8). (F and G) Immunofluorescence (F) and relative quantitative analysis (G) of the cell proliferation marker Ki67 in the intestine. Scale bar, 100 μm (n = 5). (H) Relative mRNA level of the intestinal stem cell self-renewal marker Lgr5 in the intestine (n = 8). (I) HE staining of intestinal organoids. Scale bar, 20 μm (n = 6). (J) Intestinal organoid viability (n = 6). (K) LDH levels in the medium (n = 6). (L and M) Immunofluorescence (L) and relative quantitative analysis (M) of Ki67 in intestinal organoids. Scale bar, 20 μm (n = 5). (N) Relative mRNA level of Ki67 in intestinal organoids (n = 8). (O) Relative mRNA level of Lgr5 in intestinal organoids (n = 8). The results are expressed as the median and quartile. ∗, #, and & p < 0.05; ∗∗, ##, and && p < 0.01; ∗∗∗, ###, and &&& p < 0.001 by were determined were determined by two-way ANOVA and Tukey’s post hoc test. ∗p < 0.05 compared with WT mouse Sham group; #p < 0.05 compared with WT mouse I/R group; &p < 0.05 compared with WT mouse I/R + Ficz group. I/R + Ficz group: mice were treated with daily intraperitoneal injection of 50 μg/kg Ficz for 7 consecutive days before induction of intestinal I/R; H/R + Ficz group: 200 nM Ficz was added to ILC3s and the intestinal organoid co-culture system 1 h before H/R.
Figure 7
Figure 7
IL-22 promotes the self-renewal of intestinal stem cells by activating the Wnt and Notch pathways (A and B) The relative quantitative analysis (A) and representative immunohistochemical map (B) of β-catenin expression in intestinal tissues. (C) The mRNA level of Wnt3 in intestinal tissues (n = 7–8). (D and E) The relative quantitative analysis (D) and representative immunohistochemical map (E) of Notch1 expression in intestinal tissues. (F) The mRNA level of Notch1 in intestinal tissues (n = 7–8). (G and H) The relative quantitative analysis (G) and representative immunohistochemical map (H) of β-catenin expression in intestinal organoids. (I) The mRNA level of Wnt3 in intestinal organoids (n = 7–8). (J and K) The relative quantitative analysis (J) and representative immunohistochemical map (K) of Notch1 expression in intestinal organoids. (L) The mRNA level of Notch1 in intestinal organoids (n = 7–8). The results are expressed as the median and quartile. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001 by were determined by one-way ANOVA (Tukey’s test). I/R + rmIL-22 group: mice were injected intraperitoneally with 0.25 mg/kg rmIL-22 1 h before intestinal I/R; H/R + rmIL-22 group: organoids were supplemented with 5 ng/mL rmIL-22 1 h before H/R.

References

    1. Deng F., Zhao B.C., Yang X., Lin Z.B., Sun Q.S., Wang Y.F., Yan Z.Z., Liu W.F., Li C., Hu J.J., Liu K.X. The gut microbiota metabolite capsiate promotes Gpx4 expression by activating TRPV1 to inhibit intestinal ischemia reperfusion-induced ferroptosis. Gut Microb. 2021;13:1–21. doi: 10.1080/19490976.2021.1902719. - DOI - PMC - PubMed
    1. Singer M., Deutschman C.S., Seymour C.W., Shankar-Hari M., Annane D., Bauer M., Bellomo R., Bernard G.R., Chiche J.D., Coopersmith C.M., et al. The third international consensus definitions for sepsis and septic shock (Sepsis-3) JAMA. 2016;315:801–810. doi: 10.1001/jama.2016.0287. - DOI - PMC - PubMed
    1. Rudd K.E., Johnson S.C., Agesa K.M., Shackelford K.A., Tsoi D., Kievlan D.R., Colombara D.V., Ikuta K.S., Kissoon N., Finfer S., et al. Global, regional, and national sepsis incidence and mortality, 1990-2017: analysis for the Global Burden of Disease Study. Lancet (London, England) 2020;395:200–211. doi: 10.1016/s0140-6736(19)32989-7. - DOI - PMC - PubMed
    1. Deng F., Chen Y., Sun Q.S., Lin Z.B., Min Y., Zhao B.C., Huang Z.B., Liu W.F., Li C., Hu J.J., Liu K.X. Gut microbiota dysbiosis is associated with sepsis-induced cardiomyopathy in patients: a case-control study. J. Med. Virol. 2023;95:e28267. doi: 10.1002/jmv.28267. - DOI - PubMed
    1. Dickson R.P. The microbiome and critical illness. Lancet Respir. Med. 2016;4:59–72. doi: 10.1016/s2213-2600(15)00427-0. - DOI - PMC - PubMed

Publication types