Skip to main content
Cellular and Molecular Gastroenterology and Hepatology logoLink to Cellular and Molecular Gastroenterology and Hepatology
. 2021 Aug 25;13(1):233–256. doi: 10.1016/j.jcmgh.2021.08.014

Aryl Hydrocarbon Receptor Deficiency in Intestinal Epithelial Cells Aggravates Alcohol-Related Liver Disease

Minyi Qian 1,2,3,, Jun Liu 1,2,, Danyang Zhao 1,2,, Pengpeng Cai 4,, Chuyue Pan 1,2, Wenxin Jia 1,2, Yingsheng Gao 1,2, Yufei Zhang 1,2, Nan Zhang 5, Yinan Zhang 5, Quan Zhang 6, Dalei Wu 7, Chengjie Shan 8, Meiling Zhang 8, Bernd Schnabl 9, Song Yang 10,§, Xu Shen 3,§, Lirui Wang 1,2,§,
PMCID: PMC8599170  PMID: 34454169

Abstract

Background & Aims

The ligand-activated transcription factor, aryl hydrocarbon receptor (AHR) can sense xenobiotics, dietary, microbial, and metabolic cues. Roles of Ahr in intestinal epithelial cells (IECs) have been much less elucidated compared with those in intestinal innate immune cells. Here, we explored whether the IEC intrinsic Ahr could modulate the development of alcohol-related liver disease (ALD) via the gut–liver axis.

Methods

Mice with IEC specific Ahr deficiency (AhrΔIEC) were generated and fed with a control or ethanol diet. Alterations of intestinal microbiota and metabolites were investigated by 16S ribosomal RNA sequencing, metagenomics, and untargeted metabolomics. AHR agonists were used to evaluate the therapeutic potentials of intestinal Ahr activation for ALD treatment.

Results

AhrΔIEC mice showed more severe liver injury after ethanol feeding than control mice. Ahr deficiency in IECs altered the intestinal metabolite composition, creating an environment that promoted the overgrowth of Helicobacter hepaticus and Helicobacter ganmani in the gut, enhancing their translocation to mesenteric lymph nodes and liver. Among the altered metabolites, isobutyric acid was increased in the cecum of ethanol-fed AhrΔIEC mice relative to control mice. Furthermore, both H.hepaticus and isobutyric acid administration aggravated ethanol-induced liver injury in vivo and in vitro. Supplementation with AHR agonists, 6-formylindolo[3,2-b]carbazole and indole-3-carbinol, protected mice from ALD development by specifically activating intestinal Ahr without affecting liver Ahr function. Alcoholic patients showed lower intestinal AHR expression and higher H.hepaticus levels compared with healthy individuals.

Conclusions

Our results indicate that targeted restoration of IEC intrinsic Ahr function may present as a novel approach for ALD treatment.

Keywords: Aryl Hydrocarbon Receptor, Alcohol-Related Liver Disease, Helicobacter hepaticus, Isobutyric Acid

Abbreviations used in this paper: AHR, aryl hydrocarbon receptor; Ahrfl/fl, Ahrflox/flox; AhrΔIEC, intestinal epithelial cell–specific Ahr deficiency; ALD, alcohol-related liver disease; AML12, alpha mouse liver 12; ALT, alanine aminotransferase; DMSO, dimethyl sulfoxide; FFA, fatty acid; FICZ, 6-formylindolo[3,2-b]carbazole; I3C, indole-3-carbinol; IBA, isobutyric acid; IEC, intestinal epithelial cell; LEfSe, linear discriminant analysis effect size; MLN, mesenteric lymph node; mRNA, messenger RNA; OD600, optical density analysis at 600 nm; PBS, phosphate-buffered saline; POPG, 1-palmitoyl-2-oleoyl-phosphatidylglycerol; qPCR, quantitative polymerase chain reaction; rRNA, ribosomal RNA; SCFA, short-chain fatty acid; TG, triglyceride; WT, wild-type

Graphical abstract

graphic file with name fx1.jpg


Summary.

Herein, we identified that Ahr deficiency in intestinal epithelial cells enriched Helicobacter hepaticus and Helicobacter ganmani in the gut, promoted their translocation to liver, and aggravated alcohol-related liver disease (ALD) development. Dietary supplementation with AHR agonists effectively ameliorated ALD in mice, providing a new strategy for ALD treatment.

Alcohol-related liver disease (ALD) is a leading cause of liver-related morbidity and mortality worldwide, and approximately 2 million people die of liver diseases each year, of which up to 50% mortality from cirrhosis can be attributed to alcohol.1,2 Over the past 25 years, adult per capita alcohol consumption increased by 10%.3 The exact mechanisms underpinning the pathogenesis of ALD remain unclear. Therefore, elucidating the mechanism and exploring novel therapies for ALD urgently are needed given that the current treatments are very scarce.1, 2, 3, 4

Aryl hydrocarbon receptor (Ahr) is a highly conserved, ligand-inducible transcription factor that integrates environmental, dietary, microbial, and metabolic cues to control the adaptation of multicellular organisms to environmental challenges.5,6 The Ahr is expressed in many mammalian tissues, especially in the liver, intestine, and kidney.7 In the intestine, Ahr is expressed mainly by epithelial cells and innate immune cells, and plays an important role in the regulation of innate immunity.6 For example, it regulates the number of intraepithelial lymphocytes,8 controls the production of interleukin 22 by innate lymphoid cells,9,10 and senses the bacterial virulence factors then leads to antibacterial responses.11 Compared with the role of Ahr in innate immune cells in the intestine, the function of Ahr in intestinal epithelial cells (IECs) has not been studied. One previous study showed that Ahr in IECs was associated with maintenance of epithelial barrier function.12 Given that chronic alcohol consumption can disrupt the intestinal epithelial barrier and alter gut microecology, which contributes to ALD,13,14 we investigated whether intestinal epithelial Ahr affects the progression of ALD.

Gut dysbiosis, which contributes to the pathogenesis of ALD, could present as intestinal barrier dysfunction, gut microbiota alteration, and immune system dysregulation.14, 15, 16 Bacterial components and metabolites translocate from the leaky gut through blood and lymphatics to the liver in animal models and patients with ALD.14,16,17 Once the microbial products translocate to the liver, they activate the innate immune receptors and induce the increased expression of hepatic inflammatory cytokines and lipogenesis-related factors, which promote the development of ALD.16, 17, 18 Not only bacterial products, but viable microbiota also can translocate to the liver. Using regenerating islet-derived 3 gamma (Reg3g) or Reg3b-deficient mice, we previously showed that the translocation of intestinal mucosa-associated bacteria to mesenteric lymph nodes and the liver could aggravate the progression of ALD.19 In addition, translocating Enterococcus to the liver could increase interleukin 1β secretion via the pathogen-recognition receptor Toll-like receptor 2 in Kupffer cells, resulting in hepatic inflammation and hepatocytes death,20 while administration of bacteriophages against Enterococcus faecalis significantly reduced the severity of ALD in mice.21 Translocation of gut bacteria to the liver also occurred in other liver diseases such as autoimmune hepatitis.22 Because the translocation of microbial products and intestinal bacteria play such an important role in the pathogenesis of chronic liver diseases, including ALD, it is quite essential to identify new metabolites and strains of bacteria translocating to the liver, and reveal the pathogenic mechanism by which they affect the development of ALD.

Here, we found that ethanol feeding reduced the intestinal AHR expression in mice and human beings. Compared with Ahrfl/fl mice, IEC-specific Ahr deletion mice (AhrΔIEC) showed aggravated liver injury after ethanol feeding. And the levels of Helicobacter hepaticus, Helicobacter ganmani, and isobutyric acid (IBA) were enriched in ethanol-fed AhrΔIEC mice compared with Ahrfl/fl mice. In line with the increase of IBA, bacterial gene expression of ilvE, bkdA, and pdhD, which were responsible for metabolizing valine to IBA, were increased in ethanol-fed AhrΔIEC mice. Further investigation showed that H. hepaticus and IBA could aggravate ethanol-induced liver injury in vivo and in vitro. Moreover, oral administration with agonists of AHR, 6-formylindolo[3,2-b]carbazole (FICZ)10 and indole-3-carbinol (I3C),5 could improve ethanol-induced liver damage in mice, indicating that Ahr in IECs may become a novel target for the treatment of ALD.

Results

Ahr Deficiency in IECs Aggravates Ethanol-Induced Liver Injury

We first showed that ethanol (EtOH) feeding mice subjected to the chronic-plus-binge model23 showed decreased messenger RNA (mRNA) levels of Ahr and its downstream target genes (Cyp1a1, Cyp1a2, and Cyp1b1) in IECs (Figure 1A–D). Furthermore, ethanol exposure could directly down-regulate Ahr expression in the murine intestine-derived epithelial cell line (MODE-K) (Figure 1E). To explore the effects of Ahr deficiency on the progression of alcohol-related liver disease (ALD), the AhrΔIEC mice were generated and validated by quantitative polymerase chain reaction (qPCR) and immunohistochemical staining (Figure 1F and G). Although body weight and food intake did not show significant differences between Ahrfl/fl and AhrΔIEC mice after control or ethanol feeding (Figure 1H and I), AhrΔIEC mice developed more severe liver injury and steatosis relative to Ahrfl/fl mice after ethanol feeding as shown by the increased plasma alanine aminotransferase (ALT) level, H&E staining of liver sections, Oil red O staining of liver sections, and the higher hepatic triglyceride level (Figure 1J–N). After ethanol administration, livers of AhrΔIEC mice also showed significantly higher mRNA expression of inflammation-related genes, including Il1b, Il6, Cxcl5, and Cxcl10, as compared with control mice (Figure 1O). However, we found that the hepatic mRNA levels of Tnf, Adgre1 (also known as F4/80), and Ly6g in EtOH-fed AhrΔIEC mice were comparable with those in Ahrfl/fl mice (Figure 1P). Consistently, hepatic myeloperoxidase + neutrophils and tumor necrosis factor α protein expression in AhrΔIEC mice did not differ from Ahrfl/fl mice after alcohol feeding (Figure 1Q and R). To further clarify the crucial genes contributing to the increased lipid accumulation in ethanol-fed AhrΔIEC mice, we evaluated the hepatic expression of genes involved in free fatty acids (FFAs) and triglyceride (TG) synthesis, elongation, and hydrolysis. Among these, we found that the expression of elongation of very long chain fatty acid protein 7 (Elovl7), which encodes an enzyme that is responsible for the elongation of FFAs, was increased nearly 5-fold in the livers of AhrΔIEC mice than that in Ahrfl/fl mice after ethanol administration (Figure 1O and P), suggestive of its potential role in promoting lipogenesis in AhrΔIEC mice.

Figure 1.

Figure 1

Ahr deficiency in IECs of mice exacerbates ethanol-induced liver injury. (AD) WT mice were fed a control or ethanol diet (n = 5–6 per group). (A) Body weight and food intake. (B) Plasma level of ALT. (C) Representative H&E staining images of liver sections. (D) qPCR analysis of Ahr, Cyp1a1, Cyp1a2, and Cyp1b1 in IECs isolated from control (Ctrl) and ethanol-fed mice subjected to the chronic-plus-binge model (n = 6 per group). (E) mRNA expression analysis of Ahr in murine intestine-derived epithelial cell (MODE-K) stimulated with ethanol (n = 3 independent experiments performed in 2 replicates). (F) Schematic of AhrΔIEC mice and the qPCR analysis of Ahr in IECs and lamina propria (LP) cells (n = 5 per group). (G) Representative images of immunohistochemical staining for AHR expression in small intestine of Ahrfl/fl and AhrΔIEC mice. (HN) Ahrfl/fl and AhrΔIEC mice were fed a control or ethanol diet. (H) Body weight and (I) food intake (n = 6 for Ctrl; n = 22–23 for EtOH). (J) Plasma level of ALT (n = 6 for Ctrl; n = 20–21 for EtOH). (K) Representative H&E staining images of liver sections. (L and M) Representative Oil red O–stained liver sections quantified by Image J (n = 5–6 for Ctrl; n = 8–10 for EtOH). (N) Hepatic TG content (n = 6 for Ctrl; n = 16–19 for EtOH). (O) Hepatic expression of mRNA encoded by inflammation, FFAs, and TG synthesis-related genes in ethanol-fed Ahrfl/fl and AhrΔIEC mice (n = 12–23). (P) Hepatic expression of mRNA in ethanol-fed Ahrfl/fl and AhrΔIEC mice (n = 6 per group). (Q) Myeloperoxidase staining of liver sections from ethanol-fed Ahrfl/fl and AhrΔIEC mice (n = 7–9). (R) Hepatic expression of tumor necrosis factor (TNF)α protein (n = 7–9). Scale bar: 50 μm. Data are represented as means ± SEM. ∗P < 0.05, (J and N) Mann–Whitney test; (A, B, D, E, F, H, I, M, O, P and R) unpaired t test.

To examine whether Ahr deficiency in IECs affects the absorption and metabolism of ethanol, we determined the level of ethanol in plasma and metabolism of ethanol in livers. The plasma ethanol level did not differ significantly between Ahrfl/fl and AhrΔIEC mice after ethanol feeding (Figure 2A), and the hepatic gene expression of Adh1 and Cyp2e1 (the 2 main primary enzymes that convert ethanol to acetaldehyde)24,25 were comparable between ethanol-fed Ahrfl/fl and AhrΔIEC mice (Figure 2B and C). These results indicate that Ahr deficiency in IECs promoted ethanol-induced liver disease without affecting intestinal absorption and hepatic metabolism of ethanol in mice.

Figure 2.

Figure 2

Ahr deficiency in IECs causes the progression of ethanol-induced liver damage independent of ethanol absorption and metabolism. (A) Plasma ethanol level (n = 15 for Ahrfl/fl; n = 14 for AhrΔIEC) and (B) hepatic mRNAs of Cyp2e1 and Adh1 in Ahrfl/fl and AhrΔIEC mice fed an ethanol diet (n = 21 for Ahrfl/fl; n = 20 for AhrΔIEC). (C) Western blot of hepatic CYP2E1 and its quantification (n = 3 per group). Data are represented as means ± SEM. ∗P < 0.05, unpaired t test. Ctrl, control.

Ahr Deficiency in IECs Promotes Translocation of H.hepaticus and H.ganmani to Liver in Mice

Increasing evidence has shown that intestinal microbiota dysbiosis has been implicated in the progression of ALD,14, 15, 16 we thus used 16S ribosomal RNA (rRNA) gene sequencing to investigate the effects of IEC-specific Ahr disruption on gut microbiota. As shown by the principal coordinate analysis plot, we showed that the composition of intestinal microbiota in ethanol-fed mice clustered separately from that of pair-fed control mice (Figure 3A), while the overall microbiota composition of AhrΔIEC mice did not differ significantly from Ahrfl/fl mice either on a control or ethanol diet (Figure 3A). Despite that the total number of intestinal bacteria remained unchanged between ethanol-fed Ahrfl/fl and AhrΔIEC mice (Figure 3B), the cladogram (linear discriminant analysis effect size [LEfSe] analysis) showed that the abundance of Helicobacter (the family level is Helicobacteraceae, the order level is Campylobacterales) was enriched (linear discriminant analysis score > 2) in ethanol-fed AhrΔIEC mice compared with Ahrfl/fl mice (Figure 3Ci, Cj, Ck, and D). Helicobacter also was increased slightly (P = 0.076) in control-fed AhrΔIEC mice in comparison with Ahrfl/fl mice (Figure 3E). Alistipes (the family level is Rikenellaceae) was found to be enriched in the cecal content of EtOH-fed AhrΔIEC mice as evidenced by the 16S rRNA sequencing results (Figure 3C and D), although this increase could not be confirmed using a qPCR assay (Figure 3F). In addition, the 3 main species of Alistipes, including Alistipes finegoldii, Alistipes timonensis, and Alistipes indistinctus, did not alter significantly between ethanol-fed Ahrfl/fl and AhrΔIEC mice (Figure 3F).

Figure 3.

Figure 3

Ahr deficiency in IECs promotes expansion of cecal H.hepaticus and its translocation to the liver in ethanol-induced mice. (A) Principal coordinate analysis (PCoA) plot of cecal microbiota of Ahrfl/fl and AhrΔIEC mice fed a control or ethanol diet (n = 6 for control [Ctrl]; n = 8 for EtOH). (B) Total bacteria in cecum of ethanol-fed mice (n = 18 for Ahrfl/fl; n = 16 for AhrΔIEC). (C) The cladogram of gut microbiome in different taxonomic levels from ethanol-fed mice. The taxa of different abundance in Ahrfl/fl and AhrΔIEC group are presented in blue and red, respectively (n = 8 per group). (D) Significantly altered bacterial taxa between ethanol-fed Ahrfl/fl and AhrΔIEC mice (linear discriminant analysis [LDA] score, >2 or <-2). (E) The relative abundance of Helicobacter in cecum from control-fed mice detected by 16S rRNA gene sequencing (n = 6 per group). (F) The relative levels of Alistipes and the 3 main species of Alistipes in cecum from EtOH-fed Ahrfl/fl and AhrΔIEC mice determined by qPCR (n = 6 for Ahrfl/fl; n = 5 for AhrΔIEC). (G) The relative level of H.hepaticus and H.ganmani in cecum from control or ethanol-fed mice (n = 5–6 for Ctrl; n = 18–20 for EtOH) determined by qPCR. (H and I) H.hepaticus and H.ganmani in MLNs (n = 4–6 per group) and liver (n = 8–9 per group) of ethanol-fed Ahrfl/fl and AhrΔIEC mice. (J) The relative levels of bacteria in MLNs and livers of EtOH-fed mice detected by qPCR (n = 3 per group for MLN; n = 7–9 for liver). (K) Correlation analysis between relative level of intestinal H.hepaticus with hepatic steatosis (left panel), Il6 (middle panel), and Cxcl5 expression (right panel). (L) Correlation analysis between the relative level of intestinal H.hepaticus with hepatic Il1b (upper) and Cxcl10 (lower) expression. Data are represented as means ± SEM. ∗P < 0.05, unpaired t test.

Furthermore, we also confirmed that the species of H. hepaticus and H. ganmani (2 species of Helicobacter) were increased in the cecum content of ethanol-fed AhrΔIEC mice relative to Ahrfl/fl mice as determined by qPCR (Figure 3G). Accordingly, ethanol-fed AhrΔIEC mice showed significantly higher levels of H. hepaticus and H. ganmani both in mesenteric lymph nodes (MLNs) and livers compared with the ethanol-fed Ahrfl/fl mice (Figure 3H and I), indicating that IEC-specific Ahr deficiency might facilitate the translocation of H. hepaticus and H. ganmani from the gut to liver after ethanol exposure. In addition, we also showed that levels of most commonly studied bacteria such as Enterococcus, Bifidobacterium, Clostridium, and Prevotella did not change obviously in livers between EtOH-fed Ahrfl/fl and AhrΔIEC mice, although Firmicutes (phylum) and Enterococcus (genus) were increased in MLNs of AhrΔIEC mice (Figure 3J).

More importantly, we found that the relative level of H. hepaticus in cecum was correlated positively with hepatic steatosis, Il6, and Cxcl5, but not Il1b and Cxcl10 expression (Figure 3K and L). Notably, the gene expression of gut barrier function–related proteins such as occludin (Ocln), tight junction protein 1 (Tjp1), Tjp2, and mucin 2 (Muc2) were reduced dramatically in the distal small intestine of AhrΔIEC mice relative to Ahrfl/fl mice after ethanol feeding (Figure 4A). Consistently, lipopolysaccharide levels in plasma also were increased significantly in ethanol-fed AhrΔIEC mice compared with Ahrfl/fl mice (Figure 4B), suggesting that Ahr deficiency in IECs disrupted intestinal epithelial barrier function. Of note, we failed to detect the higher mRNA levels of stem cell markers (Lrig1 and Lgr5) in the intestine of ethanol-fed AhrΔIEC mice, although previous study showed that Ahr deletion in IECs promoted stem cell proliferation upon injury of infection or chemical insults,12 the gene expression of Il22 and Il17 in the proximal small intestine did not show the obvious difference either between ethanol-fed AhrΔIEC and Ahrfl/fl mice (Figure 4C).26 Taken together, Ahr deficiency in IECs enriched intestinal Helicobacter and aggravated their translocation to liver, eventually leading to enhanced alcohol-related liver injury.

Figure 4.

Figure 4

Ahr deficiency in IECs disrupted the intestinal epithelial barrier. (A) mRNA levels of Olcn, Tjp1, Tjp2, and Muc2 in the distal small intestine (n = 5–6 per group). (B) Plasma level of lipopolysaccharide (LPS) in EtOH-fed Ahrfl/fl and AhrΔIEC mice (n = 13 for Ahrfl/fl; n = 11 for AhrΔIEC). (C) mRNA levels of leucine-rich repeats and immunoglobulin-like domains 1 (Lrig1), leucine-rich repeat containing G-protein–coupled receptor 5 (Lgr5), Il22, and Il17 in the proximal small intestine of ethanol-fed Ahrfl/fl and AhrΔIEC mice (n = 5–16), Lrig1 and Lgr5 are the stem cell markers. Data are represented as means ± SEM. ∗P < 0.05, unpaired t test. Ctrl, control.

IEC-Specific Ahr Deficiency Up-regulates IBA Level in the Intestines of Mice

To further decipher the underlying mechanism that IEC-specific Ahr deficiency promotes ethanol-induced liver damage, we also used untargeted metabolomics to assess the alteration of cecum metabolites between ethanol-fed Ahrfl/fl and AhrΔIEC mice. The top 20 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways with significant differences are shown in Figure 5A, and pathways such as protein digestion and absorption, aminoacyl-tRNA biosynthesis, and mineral absorption differed extensively between ethanol-fed AhrΔIEC and control mice. Although the tryptophan level was reduced in ethanol-fed AhrΔIEC mice, the level of kynurenic acid, which was converted from kynurenine, a main metabolite of tryptophan, was comparable between Ahrfl/fl and AhrΔIEC mice (Figure 5B). Indole and its derivatives, microbe-dependent products of tryptophan, did not alter dramatically either in these 2 groups of mice (Figure 5B). Instead, 64 metabolites with significant change were identified, among them, IBA, a short-chain fatty acid (SCFA), was increased notably in ethanol-fed AhrΔIEC mice compared with control mice (Figure 5C–E). The other SCFAs, including acetic acid, propionic acid, and butyric acid, were not different between the ethanol-fed AhrΔIEC mice and the Ahrfl/fl mice (Figure 5F). More importantly, we found that the increased level of IBA was correlated positively with hepatic steatosis and expression of inflammation-related genes (Figure 5G and H). Consistent with the increase of IBA, shotgun metagenomics analysis showed that the bacterial gene expression of ilvE, bkdA, and pdhD, which were responsible for metabolizing valine to produce IBA, were increased in the cecum of ethanol-fed AhrΔIEC mice relative to control mice (Figure 5I and J). Accordingly, we observed the reduction of valine level in these ethanol-fed AhrΔIEC mice (Figure 5CE). To explore whether alterations of gut metabolites were associated with enrichment of Helicobacter, we performed a correlation analysis between the 64 altered metabolites and the intestinal abundance of H. hepaticus or H. ganmani. The anionic phospholipid 1-palmitoyl-2-oleoyl-phosphatidylglycerol (POPG), the secondary bile acid lithocholic acid, which was reported to cause liver injury,27 nervonic acid, and the stearamide, which was increased in patients with alcoholic cirrhosis,28 showed significant positive correlations with the abundance of H. hepaticus (Figure 6A and B), while the other 4 metabolites including hypoxanthine, phenylalanine-cysteine, acamprosate, and thymine showed negative correlations with H. hepaticus (Figure 6C). In addition, we found that metabolites such as adynerin, POPG, and adenosine monophosphate were correlated positively with H. ganmani, while linoleic acid was correlated negatively with H. ganmani (Figure 6D and E).

Figure 5.

Figure 5

Ahr deficiency in IECs increases intestinal IBA. (A) The Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of intestinal metabolites from ethanol-fed Ahrfl/fl and AhrΔIEC mice (n = 6 per group). (B) Tryptophan, kynurenic acid, indole, indoleacetic acid, indoxyl acid, and indolepropionic acid levels in cecum of ethanol-fed Ahrfl/fl and AhrΔIEC mice (n = 6 per group). (C) The heatmap of the relative abundance of each significantly altered metabolite in cecum between ethanol-fed Ahrfl/fl and AhrΔIEC mice (n = 6 per group), 64 metabolites were identified with significant alteration (variable influence on projection [VIP] > 1.0; P < 0.1). (D) IBA and valine levels in the cecum content of ethanol-fed Ahrfl/fl and AhrΔIEC mice (n = 6 per group). (E) Identification of 64 metabolites with a significant increase (upward) or decrease (downward) in ethanol-fed AhrΔIEC mice compared with Ahrfl/fl mice. (F) Cecal contents of SCFAs in ethanol-fed Ahrfl/fl and AhrΔIEC mice (n = 7–10). (G) Spearman correlation of the 64 metabolites with hepatic steatosis score, ALT level, and inflammation. (H) Pearson correlation of hepatic steatosis score, Cxcl10 with intestinal IBA level. (I) Scheme for IBA-synthesis pathway. IlvE, branched-chain amino acid aminotransferase (EC:2.6.1.42); BkdA, 2-oxoisovalerate dehydrogenase E1 component (EC:1.2.4.4); and PdhD, dihydrolipoamide dehydrogenase (EC:1.8.1.4). (J) The counts of the genes involved in the IBA-synthesis pathway, including ilvE, bkdA, and pdhD, analyzed by shotgun metagenomics. n = 5 for Ahrfl/fl; n = 4 for AhrΔIEC. Data are represented as means ± SEM. ∗P < 0.05, unpaired t test.

Figure 6.

Figure 6

H.hepaticus per se could produce POPG as the metabolite that enhances H.hepaticus growth. (A) The correlation analysis between stearamide, lithocholic acid, POPG, nervonic acid, and the relative abundance of intestinal H.hepaticus. (B) Spearman rank correlation between significantly altered metabolites and the intestinal relative abundance of H.hepaticus. (C) The correlation analysis between acamprosate, thymine, hypoxanthine, phenylalanine-cysteine (Phe-Cys) and the relative abundance of intestinal H.hepaticus. (D) Spearman rank correlation between significantly altered metabolites and the intestinal relative abundance of H.ganmani. (E) The correlation analysis between adynerin, POPG, adenosine monophosphate (AMP), linoleic acid, and the relative abundance of intestinal H.ganmani (Pearson correlation). (F) POPG was observed to boost the growth of H.hepaticus in vitro. H.hepaticus was cultured anaerobically in brucella agar plates containing 8% sheep blood with or without the metabolite for 6 days. Bacteria were collected and suspended in medium and their growth were determined by OD600 measurement. n = 4 independent experiments performed in 2 replicates in each experiment. (G) The relative level of POPG in control or H.hepaticus by untargeted metabolomics (n = 2). (H) The level of POPG in the cecal content from mice fed with H.hepaticus or broth (n = 5–6). (I) Schematic representation of microinjection of H.hepaticus into the intestinal organoid. (J) POPG enhanced the growth of H.hepaticus in the intestinal organoid isolated from WT mice. H.hepaticus exposed to POPG or control medium was microinjected into intestinal organoid. After incubation for 4 days in the lumen of the organoid, bacteria were released and cultured anaerobically in brucella agar plates for 6 days and determined by OD600 analysis. n = 3 independent experiments performed in 2 replicates in each experiment. Data are represented as means ± SEM. ∗P < 0.05, unpaired t test.

Subsequently, we evaluated the effects of the correlated metabolites on the growth of H. hepaticus in vitro. We found that only the lipid POPG could significantly promote the growth of H. hepaticus (Figure 6F). Interestingly, the untargeted metabolomics analysis showed that H. hepaticus per se could produce POPG as the metabolite (Figure 6G), although we cannot exclude that POPG also possibly was derived from the host. Consistently, the intestinal POPG level in mice fed with H. hepaticus also was higher than that in control mice (Figure 6H). More importantly, we also showed that POPG could enhance H. hepaticus growth in intestinal organoids (Figure 6I and J and Supplementary Video). Therefore, we provided mechanistic evidence that POPG might be the most important metabolite to promote the enrichment of H. hepaticus in the intestine.

H.hepaticus Aggravates Ethanol-Induced Injury In Vivo and In Vitro

To further prove that the IEC-specific Ahr deficiency–induced increase of H. hepaticus promotes the development of ALD, we challenged wild-type (WT) mice with H. hepaticus every other day in a chronic-plus-binge model. H. hepaticus administration markedly increased levels of H. hepaticus (>500-fold) in feces of mice (Figure 7A). In line with this, mice gavaged with H. hepaticus developed more severe ethanol-induced hepatic injury, indicated by increased ALT level (Figure 7B), increased hepatic steatosis (Figure 7C), as well as enhancement of hepatic Cxcl5 and Cxcl10 expression (Figure 7D). Consistently, both cultured hepatocytes and Kupffer cells stimulated with H. hepaticus in vitro recapitulated the effects of H. hepaticus in vivo. H. hepaticus–treated mouse hepatocyte alpha mouse liver 12 (AML12) cells showed more lipid droplet accumulation (Figure 7E and F) and increased expression of inflammation-related genes such as Il1b, Il6, Cxcl5, and Cxcl10 in either basal or ethanol-stimulated conditions (Figure 7G). Notably, H. hepaticus–induced increment of Cxcl10 expression became more obvious in the presence of ethanol (Figure 7G). In addition, H. hepaticus also could induce the increase of Il1b, Il6, Cxcl5, and Cxcl10 gene expression in Kupffer cells (Figure 7H), which is comparable with that in hepatocytes. These results provided direct evidence that H. hepaticus could aggravate liver injury in vivo and in vitro.

Figure 7.

Figure 7

H.hepaticus promotes the progression of ALD in vivo and in vitro. (AD) Mice were gavaged with medium or H.hepaticus every other day during ethanol feeding. (A) Relative level of H.hepaticus in the feces of mice administered medium or H.hepaticus (n = 6 per group). (B) Plasma ALT level (n = 9 per group). (C) Representative H&E staining of liver sections (n = 9 per group). (D) Il1b, Il6, Cxcl5, and Cxcl10 mRNA levels in livers (n = 9 per group). (EG) AML12 cells were cultured in plates overnight, and subsequently were stimulated with medium or H.hepaticus and exposed to control (Ctrl) or EtOH simultaneously for 24 hours. n = 3–4 independent experiments performed in 2 replicates in each experiment. (E) Representative images of Oil red O staining, (F) quantified by ImageJ. (G) Il1b, Il6, Cxcl5, and Cxcl10 mRNA levels in AML12 cells. (H) Il1b, Il6, Cxcl5, and Cxcl10 mRNA levels in Kupffer cells stimulated with H.hepaticus for 4 hours. n = 4 independent experiments performed in 2 replicates in each experiment. Data are represented as means ± SEM. ∗P < 0.05, (A) Mann–Whitney test; (B, D, F, G and H) unpaired t test.

IBA Induces Liver Injury in Mice and Cultured Hepatocytes

IBA level was increased in ethanol-fed AhrΔIEC mice compared with Ahrfl/fl mice (Figure 5C), we thus investigated whether the increased IBA also could contribute to the progression of ALD. Administration of IBA in C57BL/6 mice showed obvious liver damage as indicated by the increased plasma ALT level, hepatic lipid accumulation, and higher hepatic TG level compared with control mice (Figure 8AC). We also noticed that IBA could significantly increase hepatic Cxcl5 gene expression (Figure 8D). Consistent with this, AML12 cells stimulated with IBA showed more lipid accumulation and increased gene expression of hepatic Cxcl5 and Cxcl10 (Figure 8EG). Similarly, IBA also could increase Cxcl5 and Cxcl10 expression in Kupffer cells (Figure 8H). More interestingly, we found that H. hepaticus could boost the effects of IBA on lipid accumulation (Figure 8I and J). It should be noted that IBA treatment alone or in combination with ethanol significantly up-regulated Elovl7 expression in AML12 cells (Figure 8K), which is consistent with the increased Elovl7 expression in ethanol-fed AhrΔIEC mice, which had the higher level of IBA in vivo (Figures 1O and 5CF). We thus sought to determine the effect of Elovl7 on lipid accumulation in hepatocytes. Overexpression of Elovl7 resulted in more lipid droplet accumulation in AML12 cells, either in basal or alcohol-stimulated conditions (Figure 8L and M). All of these results showed that IBA could induce hepatic steatosis and liver damage in vivo and in vitro, while H. hepaticus was able to enhance the effects of IBA on lipogenesis. We also elucidated the crucial role of Elovl7 in IBA-induced lipid accumulation.

Figure 8.

Figure 8

IBA induces liver damage in vivo and in vitro. (AD) Mice were injected intraperitoneally with vehicle or IBA for 24 or 48 hours. (A) Plasma ALT level at 24 hours (n = 8 per group). (B) Representative H&E staining of liver sections at 48 hours. (C) Hepatic TG content (n = 7–8 per group) at 48 hours. (D) Hepatic Cxcl5 mRNA at 24 hours (n = 8 per group). (E) AML12 cells were seeded in 12-well plates overnight, and were treated with control (PBS) or ethanol (100 mmol/L), and stimulated with vehicle (DMSO) or IBA (1 and 4 mmol/L) at the same time for 24 hours, and the cells were stained with Oil red O. (F) Quantification of the Oil red O staining. (G and H) mRNA levels of Cxcl5 and Cxcl10 in AML12 cells (G) treated with vehicle or IBA for 24 hours and isolated Kupffer cells, or (H) treated with vehicle or IBA for 4 hours. n = 3 independent experiments performed in 2 replicates in each experiment. (I and J) AML12 cells were seeded in 12-well plates overnight and treated with medium, IBA, H.hepaticus, and IBA+H.hepaticus for 24 hours. The cells were finally stained with Oil red O. (I) Representative images of the Oil red O staining. (J) Quantification of Oil red O staining. (K) Elovl7 mRNA levels in AML12 cells exposed to EtOH (100 mmol/L), IBA (4 mmol/L), or EtOH (100 mmol/L) + IBA (4 mmol/L). (L) Cells were transfected with empty vector or Elovl7, and exposed to control or EtOH (100 mmol/L) for 24 hours. Cells were stained with Oil red O. (M) Quantification of the Oil red O staining. For cell culture experiments, at least 3 independent experiments were performed with 2 replicates in each experiment. Scale bar: 50 μm. Data are represented as means ± SEM. ∗P < 0.05, unpaired t test. Ctrl, control.

AHR Agonists Ameliorate ALD in Mice

Two widely used agonists for AHR, I3C and FICZ,12,29 whose activation effects on Ahr were confirmed in vitro by ourselves (Figure 9A and B), were selected to explore whether supplementation with AHR agonists could be a therapeutic approach for ALD. WT mice exposed to an ethanol diet were orally gavaged with I3C (50 mg/kg) and FICZ (50 μg/kg) daily for 15 days. Although body weight and food intake showed no significant differences compared with control mice (Figure 9C), both I3C and FICZ decreased hepatic steatosis (Figure 9D), decreased liver TG levels (Figure 9E), and reduced mRNA expression of Elovl7, Il6, Cxcl5, and Cxcl10 (Figure 9F and G). Subsequently, we confirmed that I3C and FICZ activated AHR in the intestine as shown by the increased gene expression of Cyp1a1 and Cyp1b1 (Figure 9H), whereas they did not activate hepatic AHR (Figure 9I), suggesting that selectively activating intestinal AHR was sufficient to protect mice from alcohol-induced liver damage. Intriguingly, mice treated with I3C and FICZ dramatically decreased the abundance of intestinal H. hepaticus (Figure 9J). Collectively, our findings show the therapeutic potentials of AHR agonists for ALD treatment.

Figure 9.

Figure 9

AHR agonists improve ethanol-induced liver disease in mice. (A) Ahr activation by I3C (50 μmol/L) and FICZ (100 nmol/L) analyzed by luciferase assay (n = 4 independent experiments). (B) Ahr, Cyp1a1, Cyp1a2, and Cyp1b1 mRNA levels in AML12 stimulated with I3C (50 μmol/L) and FICZ (100 nmol/L) (n = 4 independent experiments). (CJ) Mice were orally administered vehicle, I3C (50 mg/kg), and FICZ (50 μg/kg), and fed an EtOH diet (n = 6–8 per group). (C) Body weight and food intake. (D) Representative liver sections stained with H&E. (E) Hepatic TG content. (F) Hepatic Elovl7 mRNA level. (G) Hepatic Il6, Cxcl5, and Cxcl10 mRNA levels. (H and I) mRNA levels of Ahr, Cyp1a1, and Cyp1b1 in distal small intestine and liver. (J) The relative level of H.hepaticus in cecum. Scale bar: 50 μm. Data are represented as means ± SEM. ∗P < 0.05, unpaired t test. Ctrl, control.

Alcoholic Patients Showed Decreased Intestinal AHR Expression and Increased H.hepaticus Level

To explore whether the decreased intestinal AHR expression was relevant to alcoholic patients, we analyzed their levels in the duodenal tissues. As expected, the mRNA and protein expression of intestinal AHR was decreased significantly in alcoholic patients compared with nonalcoholic individuals by qPCR analysis and immunohistochemical staining (Figure 10A and B). Intriguingly, compared with healthy controls, patients with alcoholic liver disease showed a higher level of fecal H. hepaticus (Figure 10C). Notably, 1 patient with an alcohol-use history for more than 30 years (2–3 bottles of liquor for daily drinking, equal to 400–600 g pure alcohol/d) and very severe aspartate aminotransferase, γ-glutamyl transferase, and Fibrosis-4 index levels, had a remarkably higher abundance of H. hepaticus in the stool sample (Figure 10C).

Figure 10.

Figure 10

Decreased intestinal AHR level and increased H.hepaticus in alcoholic patients. (A) mRNA and (B) protein levels of AHR in duodenal tissues from controls and alcoholic patients (n = 12 controls; n = 6 alcoholic patients) determined by qPCR and immunohistochemical staining. (C) The relative level of H.hepaticus in healthy controls and patients with alcoholic liver disease (n = 13 per group). (D) A schematic diagram summarizing our findings that alcohol abuse down-regulates AHR expression in IECs, which contributes to ALD progression. Alcohol depresses the expression of Ahr in IECs, and subsequently enriches intestinal H.hepaticus, H.ganmani, and IBA levels. Ahr deficiency in IECs exacerbates intestinal H.hepaticus and H.ganmani translocation through MLNs to liver via the disrupted gut barrier. POPG, 1 metabolite of H.hepaticus, in turn boosts H.hepaticus growth. H.hepaticus and IBA augment ethanol-induced liver injury by eliciting hepatic inflammation and steatosis. Supplementation with AHR agonists, FICZ and indole-3-carbinol (abundant in cruciferous vegetables), protects mice from ALD development by activating intestinal Ahr. Data are represented as means ± SEM. ∗P < 0.05, Mann–Whitney test.

Discussion

Ahr is widely expressed throughout the body, and intestinal Ahr was reported to play an important role in enteric diseases such as inflammatory bowel disease and colitis by regulation of the immune response and intestinal barrier functions.6,8,12 However, the association between intestinal Ahr and liver diseases still remains largely unexplored. The AHR ligands constitute a large family that generally can be categorized into 4 major sources: xenobiotics (eg, dioxin), dietary metabolites (eg, I3C), endogenous metabolites (eg, indole acetic acid), and microbial derivatives (eg, indirubin).30,31 Although ethanol is not identified as a direct ligand of AHR, ethanol feeding was shown to reduce microbiota-dependent AHR ligand production from tryptophan in mice such as indole acetic acid.26 Notably, AHR activation by microbial tryptophan metabolites were shown to improve ALD in mice,32 highlighting the association of ALD with intestinal AHR and microbiota. We thus attempt to explore the crosstalk between intestinal AHR and gut microbiota, as well as their metabolites, and clarify the role of intestinal AHR in the progression of ALD.

Considering that IECs function as the first line sensing intestinal environment change derived from dietary, microbial, and metabolic cues, we generated IEC-specific Ahr-deficiency mice in which Ahr expression in lamina propria cells remained unchanged. We showed that Ahr depletion in IECs did not directly affect intestinal absorption and metabolism of ethanol. In addition, we could not find that a lack of Ahr in IECs altered the intestinal mRNA levels of stem cell markers, although 1 previous study showed that Ahr deletion in IECs enhanced stem cell proliferation upon injury through infection or chemical insults.12 The gene expression of Il22 and Il17 in the proximal small intestine was not different between ethanol-fed AhrΔIEC mice and control mice.

Here, we observed that alcohol depressed the expression of Ahr in IECs in mice and human beings, and its deficiency enhanced the susceptibility to ethanol-induced liver injury. Compared with control mice, intestinal levels of Helicobacter (H. hepaticus and H. ganmani) and IBA were up-regulated in ethanol-fed AhrΔIEC mice, which were accompanied by increased intestinal permeability, and the abundance of H. hepaticus and H. ganmani in MLNs and liver also were increased accordingly in these mice. Of note, H. hepaticus per se could produce POPG as the metabolite that, in turn, enhanced the bacteria growth and promoted its translocation from gut to liver on the basis of the intestinal barrier disruption caused by Ahr deficiency. H. hepaticus and IBA aggravated ethanol-induced liver injury by eliciting hepatic inflammation and steatosis. Importantly, oral supplementation of AHR agonists FICZ and I3C (a derivative from cruciferous vegetables) markedly ameliorated ALD by activating intestinal Ahr and reducing H. hepaticus in a mouse model, suggesting a new way for the treatment of ALD (Figure 10D). Consistently, a recent study showed that the prebiotic, pectin, could improve alcohol-induced liver injury by increasing the bacterial tryptophan metabolites, and proposed that targeting intestinal AHR activation could improve alcoholic liver disease,32 however, our study, through using IEC-specific Ahr-deficiency mice, showed elaborately that it is the IEC-intrinsic Ahr that mediated the beneficial effects of AHR activation on the progression of ALD.

In this study, we showed that a specific dysbiosis, increased intestinal Helicobacter species, contributed to the progression of ALD in mice. Previously, Helicobacter species were not regarded as pathogens for human beings because the majority of colonized individuals were asymptomatic.33 However, increasing evidence has shown a strong association of intestinal Helicobacter species with enteric diseases such as Crohn’s disease and inflammatory bowel disease in patients, indicating its pathogenicity in the digestive diseases.34,35 H. hepaticus, one of the main species of Helicobacter, also could be detected in patients’ bile and liver samples, and was associated with chronic liver diseases, including primary hepatocellular carcinoma and bile duct cancer.36, 37, 38, 39 Here, we identified that the H. hepaticus level was increased markedly in patients with ALD compared with that in healthy individuals. In addition, H. ganmani, another important species of Helicobacter with increased abundance in ethanol-induced AhrΔIEC mice, was not explored in this study because of its current unavailability, however, the role of H. ganmani in ALD also deserves further investigation.

Besides the changes of gut microbiota, another explanation for the link between gut dysbiosis and liver diseases is the alteration of metabolites produced by the intestinal microbiota. Well-known metabolites such as SCFAs (eg, butyrate, acetate) could exert immunomodulatory effects both inside and outside of the intestine.40,41 Here, we showed that the SCFA IBA was increased significantly in IEC-specific Ahr-deficiency mice compared with control mice after alcohol drinking. Shotgun metagenomics analysis indicated that the pathway contributing to IBA synthesis was more activated in AhrΔIEC mice than that in Ahrfl/fl mice as evidenced by the increased bacterial gene expression of ilvE, bkdA, and pdhD. Notably, the bacterial gene ilvE, coding the enzyme responsible for catalyzing valine to 3-methyl-2-oxobutanoate, also could be found in H. hepaticus. Thus, we presumed that the increased abundance of H. hepaticus in AhrΔIEC mice also may contribute to the increased IBA. Regarding the role of IBA in liver disease, we showed that both IBA administration in vivo and treatment in cultured hepatocytes could induce liver injury when used alone or in combination with alcohol. Although we failed to detect IBA in mice serum, it still is possible that IBA could go to the liver from the portal vein to aggravate the alcohol-induced liver disease in AhrΔIEC mice given that these mice had a disrupted intestinal barrier. Our findings also are consistent with findings in nonalcoholic steatohepatitis patients containing a higher fecal IBA level.42 These results therefore suggest that fecal IBA might be developed as a new biomarker to predict the progression of ALD.

Mechanistically, we used the combination of 16S rRNA sequencing, metagenomics, and untargeted metabolomics to show that IEC-specific Ahr deficiency induced the alteration of intestinal metabolites, these metabolites, such as the anionic phospholipid POPG and the secondary bile acids lithocholic acid, nervonic acid, and stearamide (increased in patients with alcoholic cirrhosis),28 correlated positively with the abundance of H. hepaticus, while metabolites such as hypoxanthine, phenylalanine–cysteine, acamprosate, and thymine correlated negatively with the abundance of H. hepaticus. We further showed that the lipid POPG, which was produced by H. hepaticus, could in turn promote the growth of the bacteria itself. In addition, acamprosate, a drug for alcohol use disorder treatment,43,44 was identified as decreased in cecal content from EtOH-fed AhrΔIEC mice by untargeted metabolomics. It showed a negative correlation with H. hepaticus, although validation of this metabolite remains to be addressed by targeted metabolomics analysis. All of these changes in metabolite composition might directly promote the overgrowth of intestinal H. hepaticus, facilitating its translocation to the liver. At the gene level, we showed that the increased bacterial gene expression of ilvE, bkdA, and pdhD contributed to the increase of IBA in ethanol-fed AhrΔIEC mice relative to control mice, suggesting that these genes might be good targets for regulating the intestinal IBA level. More interestingly, we showed that IBA and H. hepaticus could have a synergistic effect on lipogenesis in hepatocytes. Overall, we elucidated the balance of interactions between IEC-intrinsic Ahr, gut metabolites, and microbiota, the disruption of this balance came from IEC-specific Ahr deficiency, which would lead to the alteration of gut metabolites and microbiota as well as the development of ALD in this study.

Elovl7, one enzyme responsible for the elongation of saturated FFAs, was reported to trigger lipid accumulation in differentiated adipocytes, leading to oxidative damage and inflammation.45,46 In our study, compared with Ahrfl/fl mice, we found an up-regulated gene expression of hepatic Elovl7 in ethanol-induced AhrΔIEC mice, which had more severe steatotic livers. This was consistent with the in vitro results that overexpression of Elovl7 resulted in lipid accumulation in cultured hepatocytes. Mechanistically, we identified that IBA, which was increased in AhrΔIEC mice relative to control mice, could directly up-regulate Elovl7 mRNA expression in hepatocytes. Our observations thus proved that Elovl7 might play an important role in regulating hepatic lipid accumulation in the context of alcohol-related liver disease.

In conclusion, we showed an essential role for intestinal epithelial cell intrinsic Ahr in regulating hepatic lipid accumulation and inflammation in ALD through affecting H. hepaticus and IBA levels, which might serve as predictive biomarkers for the progression of ALD. Moreover, dietary supplementation with AHR agonists provided a new therapeutic strategy for treating ALD.

Methods

Animals

Ahrfl/fl (stock no. 006203) and Villin-Cre (stock no. 021504) mice on a C57BL/6 background were purchased from Jackson Laboratory (Bar Harbor, ME) and bred in-house at China Pharmaceutical University. For IEC-specific Ahr disruption, Ahrfl/fl were crossed with Villin-Cre mice to obtain AhrΔIEC mice. WT mice on a C57BL/6 background were obtained from Beijing Vital River Laboratory Animal Technology Company (Beijing, China). All mice were housed in the specific pathogen-free facility and maintained under a temperature-controlled (22°C–23°C) room with a 12:12-hour light/dark cycle. All animal procedures were performed in compliance with the guidelines of the Institutional Animal Care and Use Committee of China Pharmaceutical University (Nanjing, China).

Animal Models

Age-matched female Ahrfl/fl and AhrΔIEC littermates (age, 8–10 wk; weight, 20–22 g) were subjected to the chronic-plus-binge model.23 In this model, 8-week-old mice (female) received a Lieber-DeCarli control liquid diet (F1259SP; Bio-Serv, Flemington, NJ) for 5 days and an ethanol liquid diet (F1258SP; Bio-Serv) for 10 days, and then were administered with a single binge of 5 g/kg ethanol (459844; Sigma-Aldrich, St. Louis, MO) on day 16. Pair-fed control mice (Ahrfl/fl and AhrΔIEC littermates) received an isocaloric substitution of dextrose diet. For AHR agonist treatment, WT female mice were gavaged daily with a volume of 100 μL vehicle (dimethyl sulfoxide [DMSO] suspended in corn oil), 50 mg/kg I3C (105220; Aladdin, Shanghai, China), or 50 μg/kg FICZ (synthetized by Professor Yinan Zhang, Nanjing University of Chinese Medicine, Nanjing, China), starting on day 1 of the binge ethanol feeding model. I3C and FICZ were dissolved in a small volume of DMSO initially, and adjusted with corn oil to prepare the final concentrations before use.12,29,47 When the mice were killed, plasma and appropriate tissues including liver, MLNs, intestine, and cecum were harvested. For IBA (I1754; Sigma-Aldrich) injection, WT female mice (age, 10 wk) were injected intraperitoneally with vehicle (phosphate-buffered saline [PBS]) or IBA (2.5% IBA diluted in PBS) at a dose of 2.5 mL/kg. Blood and liver tissues were collected at 24 or 48 hours after injection.

Human Samples

Alcoholic patients were enrolled in this study according to the pathologic examination results and inclusion criteria as described previously,21,48 and written informed consent was signed by each patient and control. All of these participates did not take any antibiotics during the 2 weeks preceding the enrolment.

For H. hepaticus abundance in feces, patients with ALD (n = 13; 12 males/1 female; mean age, 52.8 y) were enrolled in this study. Thirteen healthy volunteers (social drinkers consuming <20 g/d; 12 males/1 female; mean age, 49.07 y) were recruited as controls. Baseline features of these subjects are shown in Table 1. Fecal samples were collected, frozen immediately, and stored at −80°C. The protocol was approved by the Ethics Committee of the Beijing Ditan Hospital, Capital Medical University (Beijing, China).

Table 1.

Demographic and Clinical Parameters of Controls and Patients With Alcoholic Liver Disease

Variables Controls (n = 13) Alcoholic liver disease (n = 13)
Sex, male, n (%), n = 26 12 (92.3) 12 (92.3)
Age, y, n = 26 49.07 (33–66) 52.8 (36–78)
ALT level, U/L, n = 23 19.6 (11.9–37.4) 30.5 (6.6–83.0)
AST level, U/L, n = 23 21.5 (13.0–28.0) 61.2 (19.6–237.0)
GGT level, U/L, n = 11 259.3 (18.3–932.1)
Albumin level, g/dL, n = 13 32.5 (24.5–41.5)
Bilirubin level, mg/dL, n = 13 40.4 (13.1–92.7)
Creatinine level, mg/dL, n = 13 65.9 (34.1–123.7)
INR, n = 11 1.3 (0.83–1.79)
Platelet count, 109/L, n = 13 115.6 (24.5–402.0)
MELD, n = 11 12.8 (7.0–18.0)
FIB-4, n = 11 7.2 (0.6–21.2)
FIB-4 > 3.25, n (%) 6 (46.2)
Prior length of alcohol abuse, n = 11
10–20 y 2 (18.2%)
20–30 y 4 (36.4%)
>30 y 5 (45.4%)

ALT, alanine aminotransferase; AST, aspartate aminotransferase; FIB-4, fibrosis-4 index; GGT, γ-glutamyl transferase; INR, international normalized ratio; MELD, model for end-stage liver disease.

For intestinal AHR level analysis, duodenal tissues were collected with endoscopically normal duodenum from alcoholic patients (n = 6) and individuals without alcohol consumption (controls, n = 12). Patient characteristics are summarized in Table 2. This study protocol was approved by the Ethics Committee of the Sir Run Run Shaw Hospital, Nanjing Medical University (Nanjing, China).

Table 2.

Demographic and Clinical Parameters of Controls and Alcoholic Patients

Variables Controls (n = 12) Alcoholic patients (n = 6)
Sex, male, n (%) 6 (50.0) 6 (100.0)
Age, y 36.7 (21–59) 51.2 (37–57)
ALT level, U/L 17.7 (8.0–25.0) 22.3 (15.0–29.0)
AST level, U/L 19.3 (14.0–29.0) 17 (15.0–20.0)
GGT level, U/L 26.1 (11.0–56.0) 30.7 (18.0–44.0)
Albumin level, g/dL 44.9 (41.2–48.8) 45.4 (43.3–51.2)
Bilirubin level, μmol/L 9.4 (5.8–13.2) 8.8 (5.3–14.3)
Creatinine level, μmol/L 69.3 (58.0–79.0) 62.2 (56.0–82.0)
INR 0.97 (0.89–1.10) 0.99 (0.89–1.04)
Platelet count, 109/L 225.7 (148.0–297.0) 215.7 (173.0–335.0)
FIB-4 0.71 (0.36–1.37) 1.19 (0.52–1.71)
Prior length of alcohol abuse, n = 6
10–20 y 2 (16.7%)
20–30 y 1 (33.3%)
>30 y 3 (50.0%)

ALT, alanine aminotransferase; AST, aspartate aminotransferase; FIB-4, fibrosis-4 index; GGT, γ-glutamyl transferase; INR, international normalized ratio.

Bacterial Cultures

H.hepaticus (ATCC51449) was a gift from Professor Quan Zhang’s laboratory (Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China).49 The strain was cultured anaerobically on brucella agar (8241972; BD Difco, Detroit, MI) plates with 8% sheep blood (TX0030; Solarbio, Beijing, China) for 6 days. Bacteria collected and suspended in PBS (adjusted to optical density analysis at 600 nm [OD600] = 1.0) were used for untargeted metabolomics.

To determine the effect of metabolites for H. hepaticus growth in vitro, H. hepaticus was cultured on brucella agar plates containing 8% sheep blood with or without the metabolite. After 6 days of incubation, bacteria were collected and suspended in medium and growth was determined by OD600 measurement.

For H. hepaticus administration, WT female mice (age, 8 wk) subjected to the chronic-plus-binge model were orally gavaged every other day during the 15 days with either H. hepaticus (suspended in 200 μL broth, adjusted to OD600 = 1.0) or medium, respectively, as described.49 At the end of the experiment, mice were killed and appropriate tissues were harvested.

Biochemical Assays

Blood was harvested from the inferior caval vein of mice to tubes containing anticoagulant (0.5 mol/L EDTA-Na2), and centrifuged for 5 minutes (11,292 × g, 4°C) to obtain plasma. The levels of ALT, aspartate aminotransferase in plasma, and triglycerides in liver were determined using commercial kits (C009-2-1, C010-2-1, and A110-1-1, respectively; Nanjing Jiancheng Bioengineering Institute, Nanjing, Jiangsu, China).50

RNA Extraction and Real-Time qPCR

The total RNA from livers of mice was isolated using RNAiso Plus (9109; TaKaRa, Dalian, Liaoning, China) according to the methods described previously.50 The complementary DNA was synthesized from 1 μg RNA by using a High Capacity complementary DNA reverse-transcription kit (R312-02; Vazyme, Nanjing, Jiangsu, China). qPCR was performed as described in our previous work19,50 using SYBR Premix (Q331-02; Vazyme) according to the manufacturer’s instructions on the ABI StepOnePlus real-time PCR machine (Applied Biosystems, Foster City, CA). All qPCR primers were synthesized by Sangon Biotech (Shanghai, China) and are shown in Table 3. The relative gene expression was calculated by the 2-ΔΔCt method.

Table 3.

Primers Used for Real-Time Reverse-Transcription PCR

Name Sequence, 5’-3’
Primers for mouse
 Acaca-f GGACAGACTGATCGCAGAGAAAG
 Acaca-r TGGAGAGCCCCACACACA
 Acly-f AGCAGACATAGTCAAAGTCCAG
 Acly-r AAGATTCAGTCCCAAGTCCAAG
 Acox1-f TCGCAGACCCTGAAGAAATC
 Acox1-r CCTGATTCAGCAAGGTAGGG
 Adgre1-f CATAAGCTGGGCAAGTGGTA
 Adgre1-r GGATGTACAGATGGGGGATG
 Adh1-f AGGCATTGTTGAGAGCGTTG
 Adh1-r CGAGGCATTAGCAGATCGC
 Ahr-f CTCCTTCTTGCAAATCCTGC
 Ahr-r GGCCAAGAGCTTCTTTGATG
 Cd36-f GCGACATGATTAATGGCACA
 Cd36-r CCTGCAAATGTCAGAGGAAA
 Cpt1a-f AGTGGCCTCACAGACTCCAG
 Cpt1a-r GCCCATGTTGTACAGCTTCC
 Cxcl5-f TGATCCCTGCAGGTCCACA
 Cxcl5-r CTGCGAGTGCATTCCGCTTA
 Cxcl10-f CCAAGTGCTGCCGTCATTTTC
 Cxcl10-r GGCTCGCAGGGATGATTTCAA
 Cyp1a1-f ACCCTTACAAGTATTTGGTCGT
 Cyp1a1-r GTCATCATGGTCATAACGTTGG
 Cyp1a2-f ATCCTGGAGATCTACCGATACA
 Cyp1a2-r TATGTAGATACAGCGCTCCTTG
 Cyp1b1-f CCAAGAATACGGTCGTTTTTGT
 Cyp1b1-r GTTAGCCTTGAAATTGCACTGA
 Cyp2e1-f CACCCTCCTCCTCGTATC
 Cyp2e1-r CGCTTTGCCAACTTGGTT
 Dgat2-f CGCAGCGAAAACAAGAATAA
 Dgat2-r GAAGATGTCTTGGAGGGCTG
 Elovl1-f CCATACATCCAGATGAGGTGAA
 Elovl1-r AGCACATGACAGCCATTCAG
 Elovl5-f CCTTGAAATAGGTACTAAGTGATGC
 Elovl5-r CTCCTTCTACATCCGCCTCT
 Elovl7-f GCAATCCTCCATGAAAAAGAACT
 Elovl7-r CCAGCCTACCAGAAGTATTTGTG
 Fasn-f CTCGCTTGTCGTCTGCCT
 Fasn-r ATGTCCACACCACCAATGAG
 Il1b-f TGTGAAATGCCACCTTTTGA
 Il1b-r GGTCAAAGGTTTGGAAGCAG
 Il6-f TAGTCCTTCCTACCCCAATTTCC
 Il6-r TTGGTCCTTAGCCACTCCTTC
 Il17-f CTGTCTCTCCCGCTACTG
 Il17-r CTAGCAGCTTCCTCTGGAA
 Il22-f AGGCATTGTTGAGAGCGTTG
 Il22-r CGAGGCATTAGCAGATCGC
 Lgr5-f GGACCAGATGCGATACCGC
 Lgr5-r CAGAGGCGATGTAGGAGACTG
 Lipe-f CCTGCAAGAGTATGTCACGC
 Lipe-r GGAGAGAGTCTGCAGGAACG
 Lpl-f CTGGTGGTCCTGGGAGTTT
 Lpl-r TCCTCAGCTGTGTCTTCAGG
 Lrig1-f TTGAGGACTTGACGAATCTGC
 Lrig1-r CTTGTTGTGCTGCAAAAAGAGAG
 Ly6g-f CATCCTTCTTGTGGTCCTACTGTGTG
 Ly6g-r TCTATCTCCAGAGCAACGCAAAATCC
 Muc2-f CCTTGCAGTCAAACTCAAAGT
 Muc2-r AAGTTTGCCCCTGGCTATGAC
 Ocln-f ATTTATGATGAACAGCCCCC
 Ocln-r CATAGTCAGATGGGGGTGGA
 Srebf1-f ACAGCGGTTTTGAACGACAT
 Srebf1-r GCTCTCAGGAGAGTTGGCAC
 Tjp1-f TGCAATTCCAAATCCAAACC
 Tjp1-r AGAGACAAGATGTCCGCCAG
 Tjp2-f TTTTTGAGCTTGTTGGCTTG
 Tjp2-r GTGATTTTCTTCAACCCGGA
 Tnf-f AGATGATCTGACTGCCTGGG
 Tnf-r CTGCTGCACTTTGGAGTGAT
 18S-f AGTCCCTGCCCTTTGTACACA
 18S-r CGATCCGAGGGCCTCACTA
Primers for bacteria
 Alistipes-f ACATAGGGGGACTGAGAGGT
 Alistipes-r GCATGGCTGGTTCAGACTTG
 A finegoldii-f GTGAGGTAACGGCTCACCAA
 A finegoldii-r GCTCCTACACGTAGAAGCGT
 A Indistinctus-f GTGAGGTAACGGCTCACCAA
 A Indistinctus-r CGATACTTTCAAACAGGTACACGT
 A timonensis-f GTGAGGTAACGGCTCACCAA
 A timonensis-r CGTACTATACTTTCAGTCAGATACACG
 Bacteroidetes-f GGCGACCGGCGCACGGG
 Bacteroidetes-r GRCCTTCCTCTCAGAACCC
 Bifidobacterium-f TCGCGTCYGGTGTGAAAG
 Bifidobacterium-r CCACATCCAGCWTCCAC
 Clostridium-f ACGCTACTTGAGGAGGA
 Clostridium-r GAGCCGTAGCCTTTCACT
 Enterococcus-f AACCTACCCATCAGAGGG
 Enterococcus-r GACGTTCAGTTACTAACG
 Firmicutes-f GGAGYATGTGGTTTAATTCGA
 Firmicutes-r AGCTGACGACAACCATGCAC
 H. hepaticus-f TCGTGTCGTGAGATGTTGGG
 H. hepaticus-r TCCACCTCGCGATATTGCTC
 H. ganmani-f TGTGGAGCTTGTCTCTGCAG
 H. ganmani-r CCCAACATCTCACGACACGA
 Prevotella-f CACRGTAAACGATGGATGCC
 Prevotella-r GGTCGGGTTGCAGACC
 16S-f GTGSTGCAYGGYTGTCGTCA
 16S-r ACGTCRTCCMCACCTTCCTC

f, forward; r, reverse.

Bacterial DNA Isolation and qPCR for 16S, H.hepaticus, and H.ganmani

Bacterial genomic DNA was extracted from mice cecum content, livers, and MLNs as previously described.19,50 The bacterial abundance was quantified by qPCR, and the value of the 16S rRNA gene was normalized to cecum weight, and the relative abundance of H. hepaticus and H. ganmani in cecum, livers, and MLNs were normalized to 16S.

16S rRNA Gene Sequencing and Analysis

Cecal DNA samples from each group were selected randomly for 16S rRNA gene sequencing and analysis.19 The V4 region of the 16S rRNA gene was amplified and sequenced using the Illumina NovaSeq platform (Illumina, San Diego, CA) in Novogene Technology Co, Ltd (Beijing, China). Raw sequence data were analyzed with a pipeline51 based on USEARCH52 v10.0.240 (http://www.drive5.com/usearch) and VSEARCH53 v2.13.6 (https://github.com/torognes/vsearch). Briefly, after a combination of sequences and removal of barcodes, UNOISE354 was performed to generate amplicon sequence variants. After sequence alignment by SILVA database55 (Silva_123, http://www.drive5.com/usearch/manual/sintax_downloads.html), β-diversity was calculated with USEARCH. The principal coordinate analysis plot was generated based on Bray-Curtis distance. Then LEfSe56 (http://huttenhower.sph.harvard.edu/galaxy) and the R package edgeR57 were obtained for detecting differential taxons. All plots were drawn by R version 3.6.1 except those of LEfSe. The raw data reported here have been deposited in the National Center for Biotechnology Information Sequence Read Archive database (accession no. PRJNA663684).

Untargeted Metabolomics

Mice cecal contents were analyzed for untargeted metabolomics by using an ultrahigh performance liquid chromatography (UHPLC, 1290 Infinity LC; Agilent Technologies, Santa Clara, CA) coupled to a quadrupole time-of-flight (AB Sciex TripleTOF 6600; Applied Protein Technology, Shanghai, China). The analytes were separated on a 2.1 mm × 100 mm ACQUIY UPLC BEH 1.7-μm C18 column (Waters, Dublin, Ireland) and analyzed in both electrospray-positive and electrospray-negative ionization modes.

Metagenomics

DNA sample testing, library construction, and sequencing with an Illumina HiSeq platform were conducted at Novogene Technology Co, Ltd. The analyzing steps were described previously.51 In short, quality control was performed with Kneaddata pipeline (https://bitbucket.org/biobakery/kneaddata) to exclude the host genome. Then, these clean reads were assembled to contigs via MEGAHIT,58 and Prokka59 was used for contigs identification in a conda environment of MetaWrap,60 and CD-HIT61 was used for nonredundant genes. A gene abundance table was generated through Salmon62 and functional annotations of Kyoto Encyclopedia of Genes and Genomes (KEGG) Orthology by eggNOG-mapper63 with its related database.

SCFA Quantification

Cecum content of SCFAs were analyzed based on previous method.64 Briefly, samples (50 mg) were suspended in 200 μL distilled water and acidified with 50 μL (50%) sulfuric acid. Then, the solution was vortexed vigorously and extracted with diethyl ether. SCFAs were quantified by a gas chromatography Nexis GC-2030 (Shimadzu, Kyoto, Japan).

Staining Procedures

The liver samples were formalin-fixed and sectioned for H&E staining as described.19 Frozen liver sections were stained with Oil red O.19 Formalin-fixed intestinal samples were embedded in paraffin and stained with anti-AHR antibody (ab84833, 1:200; Abcam, Cambridge, MA) and antimyeloperoxidase antibody (ab9535, 1:200; Abcam). All sections were scanned by Nano Zoomer 2.0 RS (Hamamatsu).65

Western Blot Analysis

Western blot analysis was performed as described in our previous work.19 Briefly, intestine or liver tissues were lysed and the extracted protein was separated by sodium dodecyl sulfate–polyacrylamide gel electrophoresis, followed by transferring into a nitrocellulose membrane. The membranes were blocked and probed with antibodies against CYP2E1 (AB1252, 1:1000; Millipore, Bedford, MA) or actin (A5441, 1:5000; Sigma-Aldrich) overnight and then incubated with the corresponding secondary antibodies. The protein bands were quantified using Image J (National Institutes of Health, Bethesda, MD), normalized to actin.

Cell Culture Experiments

MODE-K cells (C495; Shanghai Hongshun Biotechnology Co, Ltd, Shanghai, China) were treated with vehicle (PBS) or ethanol (86, 100, and 200 mmol/L) for 24 hours for extraction of RNA and qPCR analysis. For H. hepaticus co-culture experiments, AML12 cells (CRL-2254; ATCC, Rockville, MD) were seeded in 12-well plates overnight, and subsequently exposed to H. hepaticus collected from the plates (OD600 = 1, suspended in cell culture medium, 100 μL/well). After incubation for 2 hours anaerobically, the plate was subjected to aerobic conditions for a total of 24 hours.66 The treated cells were stained with Oil red O or extracted RNA for qPCR analysis.

Luciferase-Reporter Assay

AML12 cells were seeded in a 24-well plate and cultured for 12 hours before transfection with Ahr reporter plasmid (kindly provided by Professor Dalei Wu, Shandong University, China)67 and pRL-TK Vector (E2241; Promega, Madison, WI). After 24 hours, cells were treated with AHR agonists (suspended in DMSO initially and diluted with medium). With another 24 hours of incubation, cells were washed with PBS and then lysed with lysis buffer (E1910; Promega). Luciferase activity was measured using a microplate reader (Tecan Spark, Männedorf, Switzerland) after adding luciferase reagent (E1910; Promega).

Isolation of IECs and Lamina Propria Cells

Small intestines were harvested immediately once mice were killed. After removal of all visible fat and Peyer’s patches, all intestines were cut open longitudinally. Then, IECs and lamina propria cells were collected based on the protocol as described previously.26

Isolation of Kupffer Cells

Kupffer cells were isolated as described in our previous work.68

Organoid Culture

Intestinal organoids were derived from WT mice and cultured as described previously.69, 70, 71 In brief, small intestine was harvested and flushed with cold Dulbecco's phosphate-buffered saline (DPBS). The intestine was cut longitudinally followed by removal of intestinal contents and villi. Intestine was washed and cut into 2- to 4-mm pieces, then tissues were collected after filtrating through a 70-μm cell strainer. After digestion in 2 mmol/L EDTA of DPBS for 30 minutes, intestine pieces were transferred into DPBS and shaken vigorously. After filtration through a 70-μm cell strainer 3 times, the crypts were harvested by centrifuging (600 × g, 5 min). Crypts were suspended in Matrigel (3432-010-01; R&D Biosystems, Minneapolis, MN) and cultured with IntestiCult OGM mouse basal medium (06000; Stemcell Technologies, Vancouver, Canada).

Microinjection of H.hepaticus Into Organoids

H.hepaticus was microinjected into the intestinal organoids as previously described.71,72 Antibiotic-free mouse basal medium was refreshed every 3 days to remove antibiotics before injection. H. hepaticus was collected and suspended in antibiotic-free medium (OD600 = 0.4). Then, H. hepaticus with or without POPG (5 μg/mL) was microinjected into lumens of organoids with FastGreen dye (0.05%, wt/vol, F7252; Sigma-Aldrich), which is used to track the injected organoids. After 2 hours, fresh medium with nonpermeant gentamicin (5 μg/mL) was added to prevent the overgrowth of bacteria outside the organoids. After 4 days of incubation, the injected organoids were dissociated with 200 μL broth containing 0.5% saponin (SAE0073; Sigma-Aldrich) for 15 minutes with repeated pipetting on ice. The bacteria were released and cultured on plates with 8% sheep blood for 6 days to determine OD600 analysis.

Synthesis of FICZ

The starting materials were 1-Boc-3-(2-ethoxyl-2-oxoethyl)-indole and 1-Boc-2-chloro-3-formyl-indole, and FICZ was synthesized as described in the previous work.73,74

Statistical Analysis

Results in this study are expressed as means ± SEM. Statistical significance between 2 groups was determined by the Mann–Whitney test or an unpaired t test. All analyses were performed using GraphPad PRISM 7.0 (San Diego, CA). P < 0.05 was considered statistically significant. For untargeted metabolomics, significance between groups was identified based on the combination of variable influence on projection values (>1.0) and P values (<0.1).

All authors had access to the study data and reviewed and approved the final manuscript.

Acknowledgments

CRediT Authorship Contributions

Minyi Qian (Data curation: Lead; Formal analysis: Lead; Funding acquisition: Supporting; Investigation: Supporting; Methodology: Equal; Writing – original draft: Lead)

Jun Liu (Data curation: Equal; Formal analysis: Equal; Investigation: Supporting; Methodology: Equal)

Danyang Zhao (Data curation: Equal; Formal analysis: Equal; Investigation: Supporting; Methodology: Equal)

Pengpeng Cai (Resources: Supporting)

Chuyue Pan (Data curation: Supporting)

wenxin Jia (Writing – original draft: Supporting)

Yingsheng Gao (Data curation: Supporting)

Yufei Zhang (Data curation: Supporting)

Nan Zhang (Resources: Supporting)

Yinan Zhang (Resources: Supporting)

Quan Zhang (Resources: Supporting)

Dalei Wu (Resources: Supporting)

Chengjie Shan (Methodology: Supporting)

Meilin Zhang (Methodology: Supporting)

Bernd Schnabl (Funding acquisition: Supporting; Supervision: Supporting; Writing – review & editing: Supporting)

Song Yang (Resources: Supporting)

Xu Shen (Resources: Supporting; Supervision: Equal)

Lirui Wang (Conceptualization: Lead; Formal analysis: Lead; Funding acquisition: Lead; Investigation: Lead; Methodology: Lead; Project administration: Lead; Supervision: Lead; Writing – original draft: Supporting; Writing – review & editing: Lead)

Footnotes

Conflicts of interest These authors disclose the following: Bernd Schnabl has consulted for Ferring Research Institute, Intercept Pharmaceuticals, HOST Therabiomics, Mabwell Therapeutics, and Patara Pharmaceuticals, and his institution (University of California San Diego) has received grant support from BiomX, NGM Biopharmaceuticals, CymaBay Therapeutics, Synlogic Operating Company, and Axial Biotherapeutics. The remaining authors disclose no conflicts.

FundingSupported by the National Natural Science Foundation of China grant 82070602 (L.W.), “Double First-Class” University Project grants CPU2018GF10 and CPU2018GY31 (L.W.), National Key Research and Development Program of China grants 2018YFC1704900 and 2018YFC1704905 (L.W.), Natural Science Foundation of Jiangsu Province grant SBK2020040688 (M.Q.), National Natural Science Foundation of China grant 31700114 (D.W.), and National Science and Technology Major Project grant 2018ZX10715-005 (S.Y.). Also supported in part by National Institutes of Health grant P30 DK120515 (B.S.).

Supplementary Material

Supplementary video

To clearly present the process that bacteria were microinjected into the intestinal organoid, Lactobacillus murinus with visible size were used instead of H.hepaticus, which is too small to be observed by video. L murinus were microinjected together with FastGreen dye (0.05%, wt/vol) to track the injected intestinal organoid.

Download video file (3MB, mp4)

References

  • 1.Avila M.A., Dufour J.F., Gerbes A.L., Zoulim F., Bataller R., Burra P., Cortez-Pinto H., Gao B., Gilmore I., Mathurin P., Moreno C., Poznyak V., Schnabl B., Szabo G., Thiele M., Thursz M.R. Recent advances in alcohol-related liver disease (ALD): summary of a Gut round table meeting. Gut. 2020;69:764–780. doi: 10.1136/gutjnl-2019-319720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Szabo G., Kamath P.S., Shah V.H., Thursz M., Mathurin P., Meeting E.-A.J. Alcohol-related liver disease: areas of consensus, unmet needs and opportunities for further study. Hepatology. 2019;69:2271–2283. doi: 10.1002/hep.30369. [DOI] [PubMed] [Google Scholar]
  • 3.Seitz H.K., Bataller R., Cortez-Pinto H., Gao B., Gual A., Lackner C., Mathurin P., Mueller S., Szabo G., Tsukamoto H. Alcoholic liver disease. Nat Rev Dis Primers. 2018;4:16. doi: 10.1038/s41572-018-0014-7. [DOI] [PubMed] [Google Scholar]
  • 4.Crabb D.W., Im G.Y., Szabo G., Mellinger J.L., Lucey M.R. Diagnosis and treatment of alcohol-associated liver diseases: 2019 practice guidance from the American Association for the Study of Liver Diseases. Hepatology. 2020;71:306–333. doi: 10.1002/hep.30866. [DOI] [PubMed] [Google Scholar]
  • 5.Kiss E.A., Vonarbourg C., Kopfmann S., Hobeika E., Finke D., Esser C., Diefenbach A. Natural aryl hydrocarbon receptor ligands control organogenesis of intestinal lymphoid follicles. Science. 2011;334:1561–1565. doi: 10.1126/science.1214914. [DOI] [PubMed] [Google Scholar]
  • 6.Rothhammer V., Quintana F.J. The aryl hydrocarbon receptor: an environmental sensor integrating immune responses in health and disease. Nat Rev Immunol. 2019;19:184–197. doi: 10.1038/s41577-019-0125-8. [DOI] [PubMed] [Google Scholar]
  • 7.Fernandez-Salguero P., Pineau T., Hilbert D.M., McPhail T., Lee S.S., Kimura S., Nebert D.W., Rudikoff S., Ward J.M., Gonzalez F.J. Immune system impairment and hepatic fibrosis in mice lacking the dioxin-binding Ah receptor. Science. 1995;268:722–726. doi: 10.1126/science.7732381. [DOI] [PubMed] [Google Scholar]
  • 8.Li Y., Innocentin S., Withers D.R., Roberts N.A., Gallagher A.R., Grigorieva E.F., Wilhelm C., Veldhoen M. Exogenous stimuli maintain intraepithelial lymphocytes via aryl hydrocarbon receptor activation. Cell. 2011;147:629–640. doi: 10.1016/j.cell.2011.09.025. [DOI] [PubMed] [Google Scholar]
  • 9.Lee J.S., Cella M., McDonald K.G., Garlanda C., Kennedy G.D., Nukaya M., Mantovani A., Kopan R., Bradfield C.A., Newberry R.D., Colonna M. AHR drives the development of gut ILC22 cells and postnatal lymphoid tissues via pathways dependent on and independent of Notch. Nat Immunol. 2011;13:144–151. doi: 10.1038/ni.2187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Qiu J., Heller J.J., Guo X., Chen Z.M., Fish K., Fu Y.X., Zhou L. The aryl hydrocarbon receptor regulates gut immunity through modulation of innate lymphoid cells. Immunity. 2012;36:92–104. doi: 10.1016/j.immuni.2011.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Moura-Alves P., Fae K., Houthuys E., Dorhoi A., Kreuchwig A., Furkert J., Barison N., Diehl A., Munder A., Constant P., Skrahina T., Guhlich-Bornhof U., Klemm M., Koehler A.B., Bandermann S., Goosmann C., Mollenkopf H.J., Hurwitz R., Brinkmann V., Fillatreau S., Daffe M., Tummler B., Kolbe M., Oschkinat H., Krause G., Kaufmann S.H. AhR sensing of bacterial pigments regulates antibacterial defence. Nature. 2014;512:387–392. doi: 10.1038/nature13684. [DOI] [PubMed] [Google Scholar]
  • 12.Metidji A., Omenetti S., Crotta S., Li Y., Nye E., Ross E., Li V., Maradana M.R., Schiering C., Stockinger B. The environmental sensor AHR protects from inflammatory damage by maintaining intestinal stem cell homeostasis and barrier integrity. Immunity. 2018;49:353–362 e5. doi: 10.1016/j.immuni.2018.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Bajaj J.S. Alcohol, liver disease and the gut microbiota. Nat Rev Gastroenterol Hepatol. 2019;16:235–246. doi: 10.1038/s41575-018-0099-1. [DOI] [PubMed] [Google Scholar]
  • 14.Sarin S.K., Pande A., Schnabl B. Microbiome as a therapeutic target in alcohol-related liver disease. J Hepatol. 2019;70:260–272. doi: 10.1016/j.jhep.2018.10.019. [DOI] [PubMed] [Google Scholar]
  • 15.Lang S., Schnabl B. Microbiota and fatty liver disease-the known, the unknown, and the future. Cell Host Microbe. 2020;28:233–244. doi: 10.1016/j.chom.2020.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Schnabl B., Brenner D.A. Interactions between the intestinal microbiome and liver diseases. Gastroenterology. 2014;146:1513–1524. doi: 10.1053/j.gastro.2014.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Chu H., Duan Y., Yang L., Schnabl B. Small metabolites, possible big changes: a microbiota-centered view of non-alcoholic fatty liver disease. Gut. 2019;68:359–370. doi: 10.1136/gutjnl-2018-316307. [DOI] [PubMed] [Google Scholar]
  • 18.Starkel P., Schnabl B. Bidirectional communication between liver and gut during alcoholic liver disease. Semin Liver Dis. 2016;36:331–339. doi: 10.1055/s-0036-1593882. [DOI] [PubMed] [Google Scholar]
  • 19.Wang L., Fouts D., Starkel P., Hartmann P., Chen P., Llorente C., DePew J., Moncera K., Ho S.B., Brenner D.A., Hooper L.V., Schnabl B. Intestinal REG3 lectins protect against alcoholic steatohepatitis by reducing mucosa-associated microbiota and preventing bacterial translocation. Cell Host Microbe. 2016;19:227–239. doi: 10.1016/j.chom.2016.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Llorente C., Jepsen P., Inamine T., Wang L., Bluemel S., Wang H.J., Loomba R., Bajaj J.S., Schubert M.L., Sikaroodi M., Gillevet P.M., Xu J., Kisseleva T., Ho S.B., DePew J., Du X., Sorensen H.T., Vilstrup H., Nelson K.E., Brenner D.A., Fouts D.E., Schnabl B. Gastric acid suppression promotes alcoholic liver disease by inducing overgrowth of intestinal Enterococcus. Nat Commun. 2017;8:837. doi: 10.1038/s41467-017-00796-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Duan Y., Llorente C., Lang S., Brandl K., Chu H., Jiang L., White R.C., Clarke T.H., Nguyen K., Torralba M., Shao Y., Liu J., Hernandez-Morales A., Lessor L., Rahman I.R., Miyamoto Y., Ly M., Gao B., Sun W., Kiesel R., Hutmacher F., Lee S., Ventura-Cots M., Bosques-Padilla F., Verna E.C., Abraldes J.G., Brown R.S., Jr., Vargas V., Altamirano J., Caballería J., Shawcross D.L., Ho S.B., Louvet A., Lucey M.R., Mathurin P., Garcia-Tsao G., Bataller R., Tu X.M., Eckmann L., van der Donk W.A., Young R., Lawley T.D., Stärkel P., Pride D., Fouts D.E., Schnabl B. Bacteriophage targeting of gut bacterium attenuates alcoholic liver disease. Nature. 2019;575:505–511. doi: 10.1038/s41586-019-1742-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Manfredo Vieira S., Hiltensperger M., Kumar V., Zegarra-Ruiz D., Dehner C., Khan N., Costa F.R.C., Tiniakou E., Greiling T., Ruff W., Barbieri A., Kriegel C., Mehta S.S., Knight J.R., Jain D., Goodman A.L., Kriegel M.A. Translocation of a gut pathobiont drives autoimmunity in mice and humans. Science. 2018;359:1156–1161. doi: 10.1126/science.aar7201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Bertola A., Mathews S., Ki S.H., Wang H., Gao B. Mouse model of chronic and binge ethanol feeding (the NIAAA model) Nat Protoc. 2013;8:627–637. doi: 10.1038/nprot.2013.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Hartmann P., Chen P., Wang H.J., Wang L., McCole D.F., Brandl K., Stärkel P., Belzer C., Hellerbrand C., Tsukamoto H., Ho S.B., Schnabl B. Deficiency of intestinal mucin-2 ameliorates experimental alcoholic liver disease in mice. Hepatology. 2013;58:108–119. doi: 10.1002/hep.26321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Lieber C.S. Relationships between nutrition, alcohol use, and liver disease. Alcohol Res Health. 2003;27:220–231. [PMC free article] [PubMed] [Google Scholar]
  • 26.Hendrikx T., Duan Y., Wang Y., Oh J.H., Alexander L.M., Huang W., Starkel P., Ho S.B., Gao B., Fiehn O., Emond P., Sokol H., van Pijkeren J.P., Schnabl B. Bacteria engineered to produce IL-22 in intestine induce expression of REG3G to reduce ethanol-induced liver disease in mice. Gut. 2019;68:1504–1515. doi: 10.1136/gutjnl-2018-317232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Alkhedaide A.Q. Preventive effect of Juniperus procera extract on liver injury induced by lithocholic acid. Cell Mol Biol (Noisy-le-Grand) 2018;64:63–68. [PubMed] [Google Scholar]
  • 28.Lian J.S., Liu W., Hao S.R., Guo Y.Z., Huang H.J., Chen D.Y., Xie Q., Pan X.P., Xu W., Yuan W.X., Li L.J., Huang J.R. A serum metabonomic study on the difference between alcohol- and HBV-induced liver cirrhosis by ultraperformance liquid chromatography coupled to mass spectrometry plus quadrupole time-of-flight mass spectrometry. Chin Med J (Engl) 2011;124:1367–1373. [PubMed] [Google Scholar]
  • 29.Wincent E., Bengtsson J., Mohammadi Bardbori A., Alsberg T., Luecke S., Rannug U., Rannug A. Inhibition of cytochrome P4501-dependent clearance of the endogenous agonist FICZ as a mechanism for activation of the aryl hydrocarbon receptor. Proc Natl Acad Sci U S A. 2012;109:4479–4484. doi: 10.1073/pnas.1118467109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Murray I.A., Patterson A.D., Perdew G.H. Aryl hydrocarbon receptor ligands in cancer: friend and foe. Nat Rev Cancer. 2014;14:801–814. doi: 10.1038/nrc3846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Stejskalova L., Dvorak Z., Pavek P. Endogenous and exogenous ligands of aryl hydrocarbon receptor: current state of art. Curr Drug Metab. 2011;12:198–212. doi: 10.2174/138920011795016818. [DOI] [PubMed] [Google Scholar]
  • 32.Wrzosek L., Ciocan D., Hugot C., Spatz M., Dupeux M., Houron C., Lievin-Le Moal V., Puchois V., Ferrere G., Trainel N., Mercier-Nome F., Durand S., Kroemer G., Voican C.S., Emond P., Straube M., Sokol H., Perlemuter G., Cassard A.-M. Microbiota tryptophan metabolism induces aryl hydrocarbon receptor activation and improves alcohol-induced liver injury. Gut. 2021;70:1299–1308. doi: 10.1136/gutjnl-2020-321565. [DOI] [PubMed] [Google Scholar]
  • 33.Chow J., Mazmanian S.K. A pathobiont of the microbiota balances host colonization and intestinal inflammation. Cell Host Microbe. 2010;7:265–276. doi: 10.1016/j.chom.2010.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Laharie D., Asencio C., Asselineau J., Bulois P., Bourreille A., Moreau J., Bonjean P., Lamarque D., Pariente A., Soulé J.C., Charachon A., Coffin B., Perez P., Mégraud F., Zerbib F. Association between entero-hepatic Helicobacter species and Crohn's disease: a prospective cross-sectional study. Aliment Pharmacol Ther. 2009;30:283–293. doi: 10.1111/j.1365-2036.2009.04034.x. [DOI] [PubMed] [Google Scholar]
  • 35.Bohr U.R., Glasbrenner B., Primus A., Zagoura A., Wex T., Malfertheiner P. Identification of enterohepatic Helicobacter species in patients suffering from inflammatory bowel disease. J Clin Microbiol. 2004;42:2766–2768. doi: 10.1128/JCM.42.6.2766-2768.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Nilsson I., Kornilovs'ka I., Lindgren S., Å Ljungh, Wadström T. Increased prevalence of seropositivity for non-gastric Helicobacter species in patients with autoimmune liver disease. J Med Microbiol. 2003;52:949–953. doi: 10.1099/jmm.0.05344-0. [DOI] [PubMed] [Google Scholar]
  • 37.Yang J., Ji S., Zhang Y., Wang J. Helicobacter hepaticus infection in primary hepatocellular carcinoma tissue. Singapore Med J. 2013;54:451–457. doi: 10.11622/smedj.2013153. [DOI] [PubMed] [Google Scholar]
  • 38.Shimoyama T., Takahashi R., Abe D., Mizuki I., Endo T., Fukuda S. Serological analysis of Helicobacter hepaticus infection in patients with biliary and pancreatic diseases. J Gastroenterol Hepatol. 2010;25(Suppl 1):S86–S89. doi: 10.1111/j.1440-1746.2010.06224.x. [DOI] [PubMed] [Google Scholar]
  • 39.Hamada T., Yokota K., Ayada K., Hirai K., Kamada T., Haruma K., Chayama K., Oguma K. Detection of Helicobacter hepaticus in human bile samples of patients with biliary disease. Helicobacter. 2009;14:545–551. doi: 10.1111/j.1523-5378.2009.00729.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Fan Y., Pedersen O. Gut microbiota in human metabolic health and disease. Nat Rev Microbiol. 2021;19:55–71. doi: 10.1038/s41579-020-0433-9. [DOI] [PubMed] [Google Scholar]
  • 41.Aron-Wisnewsky J., Vigliotti C., Witjes J., Le P., Holleboom A.G., Verheij J., Nieuwdorp M., Clément K. Gut microbiota and human NAFLD: disentangling microbial signatures from metabolic disorders. Nat Rev Gastroenterol Hepatol. 2020;17:279–297. doi: 10.1038/s41575-020-0269-9. [DOI] [PubMed] [Google Scholar]
  • 42.Da Silva H.E., Teterina A., Comelli E.M., Taibi A., Arendt B.M., Fischer S.E., Lou W., Allard J.P. Nonalcoholic fatty liver disease is associated with dysbiosis independent of body mass index and insulin resistance. Sci Rep. 2018;8:1466. doi: 10.1038/s41598-018-19753-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Addolorato G., Mirijello A., Leggio L., Ferrulli A., Landolfi R. Management of alcohol dependence in patients with liver disease. CNS Drugs. 2013;27:287–299. doi: 10.1007/s40263-013-0043-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Leggio L., Lee M.R. Treatment of alcohol use disorder in patients with alcoholic liver disease. Am J Med. 2017;130:124–134. doi: 10.1016/j.amjmed.2016.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Shin D., Shin J.Y., McManus M.T., Ptácek L.J., Fu Y.H. Dicer ablation in oligodendrocytes provokes neuronal impairment in mice. Ann Neurol. 2009;66:843–857. doi: 10.1002/ana.21927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Kihara A. Very long-chain fatty acids: elongation, physiology and related disorders. J Biochem. 2012;152:387–395. doi: 10.1093/jb/mvs105. [DOI] [PubMed] [Google Scholar]
  • 47.Singh N.P., Singh U.P., Rouse M., Zhang J., Chatterjee S., Nagarkatti P.S., Nagarkatti M. Dietary indoles suppress delayed-type hypersensitivity by Inducing a switch from proinflammatory Th17 cells to anti-inflammatory regulatory T cells through regulation of microRNA. J Immunol. 2016;196:1108–1122. doi: 10.4049/jimmunol.1501727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Brandl K., Hartmann P., Jih L.J., Pizzo D.P., Argemi J., Ventura-Cots M., Coulter S., Liddle C., Ling L., Rossi S.J., DePaoli A.M., Loomba R., Mehal W.Z., Fouts D.E., Lucey M.R., Bosques-Padilla F., Mathurin P., Louvet A., Garcia-Tsao G., Verna E.C., Abraldes J.G., Brown R.S., Jr., Vargas V., Altamirano J., Caballería J., Shawcross D., Stärkel P., Ho S.B., Bataller R., Schnabl B. Dysregulation of serum bile acids and FGF19 in alcoholic hepatitis. J Hepatol. 2018;69:396–405. doi: 10.1016/j.jhep.2018.03.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Cao S., Zhu C., Feng J., Zhu L., Yin J., Xu Y., Yang H., Huang Y., Zhang Q. Helicobacter hepaticus infection induces chronic hepatitis and fibrosis in male BALB/c mice via the activation of NF-κB, Stat3, and MAPK signaling pathways. Helicobacter. 2020;25 doi: 10.1111/hel.12677. [DOI] [PubMed] [Google Scholar]
  • 50.Qian M., Hu H., Yao Y., Zhao D., Wang S., Pan C., Duan X., Gao Y., Liu J., Zhang Y., Yang S., Qi L.W., Wang L. Coordinated changes of gut microbiome and lipidome differentiates nonalcoholic steatohepatitis (NASH) from isolated steatosis. Liver Int. 2020;40:622–637. doi: 10.1111/liv.14316. [DOI] [PubMed] [Google Scholar]
  • 51.Liu Y.X., Qin Y., Chen T., Lu M., Qian X., Guo X., Bai Y. A practical guide to amplicon and metagenomic analysis of microbiome data. Protein Cell. 2021;12:315–330. doi: 10.1007/s13238-020-00724-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Edgar R.C. Search and clustering orders of magnitude faster than BLAST. Bioinformatics. 2010;26:2460–2461. doi: 10.1093/bioinformatics/btq461. [DOI] [PubMed] [Google Scholar]
  • 53.Rognes T., Flouri T., Nichols B., Quince C., Mahe F. VSEARCH: a versatile open source tool for metagenomics. PeerJ. 2016;4 doi: 10.7717/peerj.2584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Edgar R.C. UNOISE2: improved error-correction for Illumina 16S and ITS amplicon sequencing. bioRxiv. 2016 doi: 10.1101/081257. [DOI] [Google Scholar]
  • 55.Yilmaz P., Parfrey L.W., Yarza P., Gerken J., Pruesse E., Quast C., Schweer T., Peplies J., Ludwig W., Glöckner F.O. The SILVA and “All-species Living Tree Project (LTP)” taxonomic frameworks. Nucleic Acids Res. 2013;42:D643–D648. doi: 10.1093/nar/gkt1209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Segata N., Izard J., Waldron L., Gevers D., Miropolsky L., Garrett W.S., Huttenhower C. Metagenomic biomarker discovery and explanation. Genome Biol. 2011;12:R60. doi: 10.1186/gb-2011-12-6-r60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Robinson M.D., McCarthy D.J., Smyth G.K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–140. doi: 10.1093/bioinformatics/btp616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Li D., Liu C.M., Luo R., Sadakane K., Lam T.W. MEGAHIT: an ultra-fast single-node solution for large and complex metagenomics assembly via succinct de Bruijn graph. Bioinformatics. 2015;31:1674–1676. doi: 10.1093/bioinformatics/btv033. [DOI] [PubMed] [Google Scholar]
  • 59.Seemann T. Prokka: rapid prokaryotic genome annotation. Bioinformatics. 2014;30:2068–2069. doi: 10.1093/bioinformatics/btu153. [DOI] [PubMed] [Google Scholar]
  • 60.Uritskiy G.V., DiRuggiero J., Taylor J. MetaWRAP-a flexible pipeline for genome-resolved metagenomic data analysis. Microbiome. 2018;6:158. doi: 10.1186/s40168-018-0541-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Fu L., Niu B., Zhu Z., Wu S., Li W., CD-HIT accelerated for clustering the next-generation sequencing data. Bioinformatics. 2012;28:3150–3152. doi: 10.1093/bioinformatics/bts565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Patro R., Duggal G., Love M.I., Irizarry R.A., Kingsford C. Salmon provides fast and bias-aware quantification of transcript expression. Nat Methods. 2017;14:417–419. doi: 10.1038/nmeth.4197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Huerta-Cepas J., Forslund K., Coelho L.P., Szklarczyk D., Jensen L.J., von Mering C., Bork P. Fast genome-wide functional annotation through orthology assignment by eggNOG-Mapper. Mol Biol Evol. 2017;34:2115–2122. doi: 10.1093/molbev/msx148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Zhang S., Wang H., Zhu M.J. A sensitive GC/MS detection method for analyzing microbial metabolites short chain fatty acids in fecal and serum samples. Talanta. 2019;196:249–254. doi: 10.1016/j.talanta.2018.12.049. [DOI] [PubMed] [Google Scholar]
  • 65.Wang L., Mazagova M., Pan C., Yang S., Brandl K., Liu J., Reilly S.M., Wang Y., Miao Z., Loomba R., Lu N., Guo Q., Liu J., Yu R.T., Downes M., Evans R.M., Brenner D.A., Saltiel A.R., Beutler B. YIPF6 controls sorting of FGF21 into COPII vesicles and promotes obesity. Proc Natl Acad Sci U S A. 2019;116:15184–15193. doi: 10.1073/pnas.1904360116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Long X., Wong C.C., Tong L., Chu E.S.H., Ho Szeto C., Go M.Y.Y., Coker O.O., Chan A.W.H., Chan F.K.L., Sung J.J.Y., Yu J. Peptostreptococcus anaerobius promotes colorectal carcinogenesis and modulates tumour immunity. Nat Microbiol. 2019;4:2319–2330. doi: 10.1038/s41564-019-0541-3. [DOI] [PubMed] [Google Scholar]
  • 67.Wu D., Potluri N., Kim Y., Rastinejad F. Structure and dimerization properties of the aryl hydrocarbon receptor PAS-A domain. Mol Cell Biol. 2013;33:4346–4356. doi: 10.1128/MCB.00698-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Mazagova M., Wang L., Anfora A.T., Wissmueller M., Lesley S.A., Miyamoto Y., Eckmann L., Dhungana S., Pathmasiri W., Sumner S., Westwater C., Brenner D.A., Schnabl B. Commensal microbiota is hepatoprotective and prevents liver fibrosis in mice. FASEB J. 2015;29:1043–1055. doi: 10.1096/fj.14-259515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Qu M., Xiong L., Lyu Y., Zhang X., Shen J., Guan J., Chai P., Lin Z., Nie B., Li C. Establishment of intestinal organoid cultures modeling injury-associated epithelial regeneration. Cell Res. 2021;31:259–271. doi: 10.1038/s41422-020-00453-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Sato T., Vries R.G., Snippert H.J., van de Wetering M., Barker N., Stange D.E., van Es J.H., Abo A., Kujala P., Peters P.J., Clevers H. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459:262–265. doi: 10.1038/nature07935. [DOI] [PubMed] [Google Scholar]
  • 71.Pleguezuelos-Manzano C., Puschhof J., Rosendahl Huber A., van Hoeck A. Mutational signature in colorectal cancer caused by genotoxic pks(+) E. coli. Nature. 2020;580:269–273. doi: 10.1038/s41586-020-2080-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Bartfeld S., Bayram T., van de Wetering M., Huch M., Begthel H., Kujala P., Vries R., Peters P.J., Clevers H. In vitro expansion of human gastric epithelial stem cells and their responses to bacterial infection. Gastroenterology. 2015;148:126–136.e6. doi: 10.1053/j.gastro.2014.09.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Sekine D., Okeda S., Hosokawa S. Concise synthesis of 6-formylindolo[3,2-b]carbazole (FICZ) Chem Lett. 2014;43:1932–1934. [Google Scholar]
  • 74.Bergman JTaJ. Synthesis of 6-formylindolo[3,2-b]carbazole, an extremely potent ligand for the aryl hydrogen (Ah) receptor. Tetrahedron Lett. 1998;39:1619–1622. [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary video

To clearly present the process that bacteria were microinjected into the intestinal organoid, Lactobacillus murinus with visible size were used instead of H.hepaticus, which is too small to be observed by video. L murinus were microinjected together with FastGreen dye (0.05%, wt/vol) to track the injected intestinal organoid.

Download video file (3MB, mp4)

Articles from Cellular and Molecular Gastroenterology and Hepatology are provided here courtesy of Elsevier

RESOURCES