Secretory Ig A (sIgA) plays an important role in the maintenance of intestinal homeostasis via cross-talk with gut microbiota. The defects in sIgA production could elicit dysbiosis of commensal microbiota and subsequently facilitate the development of inflammatory bowel disease. Our previous study revealed activating transcription factor 3 (ATF3) as an important regulator of follicular helper T (TFH) cells in gut. ATF3 deficiency in CD4+ T cells impaired the development of gut TFH cells, and therefore diminished sIgA production, which increased the susceptibility to murine colitis. However, the potential role of microbiota in ATF3-mediated gut homeostasis remains incompletely understood. In this study, we report that both Atf3−/− and CD4creAtf3fl/fl mice displayed profound dysbiosis of gut microbiota when compared with their littermate controls. The proinflammatory Prevotella taxa, especially Prevotella copri, were more abundant in ATF3-deficient mice when compared with littermate controls. This phenotype was obviously abrogated by adoptive transfer of either TFH cells or IgA+ B cells. Importantly, depletion of gut microbiota dramatically alleviated the severity of colitis in Atf3−/− mice, whereas transfer of microbiota from Atf3−/− mice to wild-type recipients increased their susceptibility to colitis. Collectively, these observations indicate the importance of IgA-microbiota interaction in ATF3-mediated gut homeostasis.

Immunoglobulin A represents the most abundant Ig in gut, which plays a critical role in the maintenance of intestinal homeostasis (1). Secretory IgA (sIgA) is constitutively released into the lumen by intestinal epithelium, where it prevents the invading pathogens from binding to mucosal epithelial cells and neutralizes the toxins (2). There are two distinct manners to produce sIgA: T cell–independent and T cell–dependent mechanisms (3, 4). Under homeostasis, sIgA is dominantly produced through T cell–independent manner and displays lower affinity. Once invading enteric pathogens are recognized by the gut immune system, high-affinity IgA is produced via T cell–dependent manner. Follicular helper T (TFH) cells in Peyer patches (PPs) and mesenteric lymphoid nodes play a critical role in T cell–dependent IgA production through facilitating the somatic hypermutation process in germinal center (GC) B cells (5, 6). Mice and human with defective IgA production displayed enhanced susceptibility to inflammatory bowel disease (1).

It has been well documented that the gut microbiota could shape hosts’ immune system to prevent inflammation (7, 8). The symbiotic bacteria promote immune tolerance, and the pathogenic ones trigger intestinal inflammation such as inflammatory bowel disease (9). sIgA has been reported to regulate the composition of bacteria and affects its motility, thus preventing the colonization of pathogenic bacteria and promoting the integrity of gut mucosal barrier (10). Meanwhile, the interaction between the microbiota and gut immune system could stimulate the secretion of IgA into lumen (11). Therefore, the symbiotic relationship between microbiota and intestinal immune system is essential to prevent inflammation and disorders in gut.

Activating transcription factor 3 (ATF3) is an early inducible gene in response to stress or environmental stimuli (12). ATF3 is a basic leucine zipper (bZip) transcription factor and it can activate or inhibit the transcription of its target genes through binding to the canonical ATF/CREB site or the similar AP-1 site (13). Regulation of immune responses by ATF3 has been reported in distinct immune cell types. ATF3 could restrict the inflammatory responses by suppressing the expression of TLR4 (14) or CCL4 in macrophage (15). The importance of ATF3 in inflammatory disorders including, but not limited to, asthma (16), sepsis (17), autoimmune diseases (18, 19) has been recognized. Recently, we and other group reported that ATF3 plays an important role in the maintenance of gut homeostasis. Deficiency of ATF3 significantly enhanced the susceptibility of chemically induced colitis in mice (20, 21). The defective generation of TFH cells in PPs and the resulted impaired IgA production contributed to the aggravated colitis in ATF3-deficient mice (21). However, it remains to be clarified that whether the microbiota participates in the aggravated colitis observed in ATF3-deficient mice.

In this study, a significant difference in the composition and diversity of gut microbiota between ATF3-deficient and ATF3-sufficient mice was observed. Particularly, Prevotella was notably increased in ATF3-deficient mice. Adoptive transfer of TFH cells or IgA+ B cells abrogated these differences. Further microbiota depletion and fecal transplantation experiments showed that microbiota from ATF3-deficient mice significantly aggravated colitis in mice. These observations revealed a critical role of microbiota in ATF3-mediated gut homeostasis.

Atf3fl/fl mice, CD4cre mice, and CD4creAtf3fl/fl mice were previously described (21). Atf3−/− mice were kindly provided by Dr. T. Hai (Ohio State University, Columbus, OH). C57BL/6 mice were from the Laboratory Animal Center of Sun Yat-sen University. C57BL6/Ly5.1 (CD45.1)–congenic mice and Rag1−/− mice were purchased from Shanghai Nanfang Research Center for Model Organisms (Shanghai, China). All mice were maintained on a C57BL/6 background and kept in specific pathogen–free facilities; sex- and age-matched littermates were used as controls. Experiments in this study were performed in accordance with the Institutional Animal Care and Use Committee of Sun Yat-sen University.

For dextran sulfate sodium (DSS)–induced colitis, mice were exposed to 2.5% (w/v) DSS (MP Biomedicals) in drinking water for 7 d, followed by 2 d with normal water. Mice were examined daily for body weight and disease activity index (DAI) to evaluate the severity of colitis. DAI and colitis score were recorded as previous reports (21, 22).

T cell–mediated colitis model was performed as previous report (23). Briefly, spleen and s.c. lymph nodes from 6 to 8 wk C57BL/6J mice were digested into single-cell suspension, RBCs were depleted, and CD4+ T cells were enriched using Mouse CD4 T Lymphocyte Enrichment Set (catalog no. 55813; BD Biosciences). Then CD4+ T cells were stained with anti-CD4, anti-CD25, and anti-CD45RB. A fraction of the CD4+CD25CD45RBhi population was sorted using an FACS Aria III (BD Biosciences). The purity of the sorted cells was higher than 98%, as evaluated by flow cytometric analysis. Rag1−/− mice were then i.p. injected with 0.4 × 106 naive CD4 T cells in 200 μl of sterile PBS. The severity of colitis was monitored weekly.

PPs were carefully dissected from the small intestine under a stereo microscope using fine scissors. For immunofluorescence staining, PPs were embedded in OCT. Frozen sections were prepared on microslides and stored at −70°C until use. For flow cytometric analysis and sorting, PPs were digested in RPMI-1640 medium (Invitrogen) containing 100 μg/ml Liberase TM and 50 μg/ml DNase I (Sigma-Aldrich) at 37°C on a roto-mixer. After 15 min, tissues were mixed using a 1-ml pipette. The supernatant was collected and resuspended in ice-cold MACS buffer (pH 7.4; PBS plus 2% FBS and 2 mM EDTA). The remaining tissues were digested again in fresh digestion buffer, and supernatant was collected after 15 min. Supernatants from the two steps were combined and passed through 70-μm filters, and cells were collected for further analysis.

For the depletion of intestinal microbiota, wild-type (WT) or Atf3−/− mice were administered with an antibiotic mixture (a mixture of 1 g/l ampicillin, 0.5 g/l vancomycin, 1 g/l neomycin sulfate, and 1 g/l metronidazole [AVNM]) in drinking water for 4 wk. For Rag1−/− mice, 2-wk treatment of AVNM was applied to deplete microbiota. The efficiency of depletion was evaluated by PCR detection of 16S rRNA gene copy numbers (total eubacteria) based on bacteria per gram feces (24, 25).

For fecal transplantation, the procedure started 12 h after the withdrawal of antibiotics. Fresh intestinal contents from WT or Atf3−/− mice were collected, weighed, and suspended in sterile PBS (200 mg in 1 ml of PBS). The solution was vortexed for 10 s, followed by centrifuge at 800 × g for 3 min to remove the large particles. Then the supernatants were collected and transplanted into the AVNM-treated mice by gavage every 3 d with 200 μl per mouse. PCR detection of 16S rRNA genes was performed to evaluate the efficiency of fecal transplantation (26, 27). The recipients were then exposed to DSS to induce colitis. In T cell–mediated colitis model, Rag1−/− mice were colonized with feces from either WT or Atf3−/− mice every 3 d for 8 wk after depletion of intestinal microbiota.

For analysis of IgA-coated bacteria, we followed a previous study (28). Briefly, bacterial pellets were washed twice by resuspending in 500 μl of sterile PBS and spinning for 5 min at 8000 × g. The suspension was then blocked with 500 μl of sterile PBS containing 1% BSA for 15 min. Samples were spun at 8000 × g for 5 min and stained with rat anti-mouse IgA Ab (catalog no. 1040-20; Southern Biotech) for 30 min at 4°C. Pellets were washed twice in sterile PBS and analyzed on an LSR Fortessa Flow Cytometer (BD Biosciences).

Fecal DNA was extracted using Power Fecal DNA Isolation Kit (MO BIO). Amplification of the V1-V3 region of the 16S rRNA gene was performed as previously described (29). Briefly, each reaction mixture contains 50 ng of purified DNA, 0.2 mM dNTPs, 1.5 mM MgCl2, 2.5 U of Platinum Taq DNA polymerase, 2.5 μl of 10× PCR buffer, and 0.5 μM of each primer designed to amplify the V1-V3: V1_27F (5′-AGAGTTTGATCCTGGCTCAG-3′) and V3_534R (5′-GCATTACCGCGGCTG CTGG-3′). A unique 12-base Golay barcode (Ns) preceded the primers to allow sample identification, and one to eight additional nucleotides were placed in front of the barcode to offset the sequencing of the primers. Amplicons were purified using the QIAquick PCR Purification Kit (Qiagen). PCR products were quantified and pooled at equimolar amounts before ligation of Illumina barcodes and adaptors, according to Illumina TruSeq Sample Preparation protocol. The completed library was sequenced on an Illumina MiSeq platform following the Illumina-recommended procedures. For data analysis, reads were preprocessed, and quality control was performed by FastQC. QIIME (Version 1.9.1) was used to handle operational taxonomic unit (OTU), sequences with ≥97% similarity were assigned to the same OTUs, and the representative sequence for each OTU was screened for further taxonomy assignment using the Ribosomal Database Project (Release 11). Data analysis of α diversity and β diversity was performed on QIIME, the visualization was displayed by R software (Version 3.3.1).

Fecal genomic DNA was extracted by the Power Fecal DNA Isolation Kit (MO BIO). Bacterial genomic DNA was quantified by eubacteria-specific 16S primers. The abundance of the indicated bacteria was analyzed with a SYBR Premix ExTaq Kit (Takara Bio) at CFX connect Optics Module (Bio-Rad Laboratories). The relative abundance was calculated by comparing the cycle threshold values using the 2−ΔΔCycle threshold method. The primer sequences for bacteria used were listed: Universal 16S (5′-ACTCCTACGGGAGGCAGCAGT-3′ and 5′-ATTACCGCGGCTGCTGGC-3′) (26), Lactobacillus (5′-TGGAAACAGRTGCTAATACCG-3′ and 5′-GTCCATTGTGGAA GATTCCC-3′) (30), Prevotella (5′-CACCAAGGCGACGATCA-3′ and 5′-GGATA ACGCCYGGACCT-3′) (31), Clostridium XIVa (5′-AAATGACGGTACCTGACT AA-3′ and 5′-CTTTGAGTTTCATTCTTGCGAA-3′) (31), Bacteroides spp (5′-CGAT GGATAGGGGTTCTGAGAGGA-3′ and 5′-GCTGGCACGGAGTTAGCCGA-3′) (31), Akkermansia (5′-CAGCACGTGAAGGTGGGGAC-3′, 5′-CCTTGCGGTTGGC TTCAGAT-3′) (31), Prevotellaceae (5′-CCAGCCAAGTAGCGTGCA-3′ and 5′-TG GACCTTCCGTATTACC-3′) (32), Prevotella copri (5′-CCGGACTCCTGCCCCTGCAA-3′ and 5′-GTTGCGCCAGGCACTGCGAT-3′) (26), Prevotella falsenii (5′-CGTGGACCAAA GTTATTTCGGTAGA-3′ and 5′-AAACAACCCCTCATTTCTCA-3′) (33), Prevotella intermedia (5′-AATACCCGATGTTGTCCACA-3′ and 5′-TTAGCCGGTCCTT ATTCGAA-3′) (34), and Prevotella ruminicola (5′-GAAAGTCGGATTAATGCTCTATGTTG-3′ and 5′-CATCCTA TAGCGGTA AACCTTTGG-3′) (35).

Single-cell suspensions were blocked with anti-CD16/CD32 before staining with fluorochrome-conjugated Abs. Staining of surface markers was performed in PBS containing 2% BSA. For intracellular staining, cells were fixed with the Transcription Factor Staining Buffer Set (eBioscience) after staining with Abs against surface markers. Staining of Abs against specific transcription factors was performed in 1× permeabilization buffer (eBioscience) for 1 h at room temperature. To detect cytokine production, cells were stimulated with 50 ng/ml PMA, 1 μg/ml ionomycin, and 2 μg/ml brefeldin A for 4 h at 37°C before analysis. The Abs used were listed below: CD4 (GK1.5; eBioscience), CD25 (PC61; BioLegend), CD45RB (C363-16A; BioLegend), CD45.1 (A20; BioLegend), CD45.2 (104; eBioscience), CXCR5-biotin (2G8 (RUO); BD Bioscience), streptavidin (eBioscience), PD-1 (J43; eBioscience), ICOS (7E.16G9; eBioscience), Bcl6 (K112-91; BD Bioscience), B220 (RA-3-6B2; BioLegend), IgA (1040-20; Southern Biotech), CD95 (Fas) (15A7; eBioscience), GL7 (GL-7; eBioscience), PNA (Sigma-Aldrich), CD138 (281-2; BioLegend), Foxp3 (FJK-16S; eBioscience), recombinant mouse IL-21R Fc chimera (596-MR; R&D Systems), R-PE–conjugated anti–human IgG (109-116-098; Jackson ImmunoResearch), Ki67 (16A8; BioLegend), and annexin V (556420; BD Bioscience). Fluorochrome-labeled cells were acquired on an LSR Fortessa Flow Cytometer (BD Biosciences). Data were analyzed with FlowJo V10.0.7 software (FlowJo, Ashland, Oregon).

The p values were calculated using unpaired two-tailed Student t tests in GraphPad Prism. A p value <0.05 is considered statistically significant. The p values are as follows: *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001.

The following methods, including H&E staining, immunofluorescence staining, and ELISA, were performed as previously reported (21).

Consistent with previous report, whole-body knockout of ATF3 (Atf3−/−) led to aggravated colitis when the mice were challenged with 2.5% DSS, as revealed by increased weight loss, DAI, colon shortening, as well as colitis score (Supplemental Fig. 1A–F). When mice were challenged with higher dose of DSS (3%), Atf3−/− mice displayed a dramatically lower chances survival than WT control (Supplemental Fig. 1G). In agreement with the aggravated colitis, PPs from Atf3−/− mice were severely shrunken in size (Supplemental Fig. 2A), and their cellularity was only approximately half of those from WT littermates (Supplemental Fig. 2B). Consistent with the previous report (21), the frequency of TFH cells in PPs from Atf3−/− mice, as well as their secretion of IL-21 cytokine, were pronouncedly reduced when compared with those of WT control (Supplemental Fig. 2C). No significant differences were observed in the level of regulatory TFH cells (Supplemental Fig. 2D). PP TFH cells from Atf3−/− mice displayed lower expression of TFH markers (including Bcl6, PD-1, and ICOS) when compared with ATF3-sufficient TFH cells (Supplemental Fig. 2E). The reduction of PP TFH cells in Atf3−/− mice was not caused by the changes in cell proliferation or cell apoptosis, as represented by Ki-67 staining or annexin V staining, respectively (Supplemental Fig. 2F, 2G). As a consequence of the impaired TFH cells, GC reaction was apparently diminished in PPs from Atf3−/− mice, as evaluated by flow cytometric analysis of GC B cells and immunofluorescence staining of GC area (Supplemental Fig. 3A, 3B). The defects in TFH/GC B axis subsequently reduced IgA+ B cells in PPs, as well as the levels of IgA+ plasma cells in lamina propria of the small intestine and colon in Atf3−/− mice (Supplemental Fig. 3C–E). The lower amounts of intestinal IgA in Atf3−/− mice were further confirmed by ELISA (Supplemental Fig. 3F). It is important to point out that under steady-state conditions PPs from ATF3-deficient mice also showed pronounced lower cellularity when compared with WT control (data not shown). The reduction of PP TFH cells as well as diminished GC reaction and IgA production were also observed in Atf3−/− mice in the absence of DSS (data not shown). Collectively, these observations indicate that whole-body knockout of ATF3 impaired the TFH/GC B/IgA axis in gut.

Intestinal IgA plays a critical role in the regulation of the composition and diversity of microbiota, therefore promoting immune homeostasis in gut (36). We next sought to investigate whether the impaired TFH/IgA axis in gut impacts intestinal microbiota in Atf3−/− mice. It was found that the proportion of IgA-bound microbiota was substantially diminished in Atf3−/− mice when compared with WT littermates (Fig. 1A). High-throughput 16S rRNA sequencing of fecal DNA showed that the diversity of gut microbiota of Atf3−/− mice was significantly lower when compared with WT controls, as indicated by the numbers of OTU (Fig. 1B, 1C). Principal coordinates analysis of the β (between-sample) diversity revealed a distinct composition of microbial community between Atf3−/− mice and WT controls (Fig. 1D). Further Shannon index showed that the richness and biodiversity of species were significantly reduced in Atf3−/− mice (Fig. 1E). Evaluation of relative abundance of distinct phylum showed that Firmicutes was obviously increased, whereas Bacteriodetes was pronouncedly decreased in Atf3−/− mice (Fig. 1F). Interestingly, colitogenic Prevotella (26) and Akkermansia (37) were much more abundant, whereas intestinal Lactobacillus (30) was less abundant in Atf3−/− mice, as indicated by heatmap depicting (Fig. 1G), and percentage analysis (Fig. 1H), in genus level. These observations indicate that ATF3 deficiency causes dysbiosis of gut microbiota.

To confirm the changes in microbiota composition, quantitative PCR (qPCR) analysis of 16S rRNA genes were next performed. The relative abundance of colitogenic taxa Prevotella and Bacteriodes were remarkably elevated, whereas Lactobacillus taxa were dramatically decreased in Atf3−/− mice when compared with WT littermates (Fig. 2A). DNA electrophoresis confirmed the dramatic changes of Prevotella and Lactobacillus in Atf3−/− mice (Fig. 2B). Moreover, Prevotellaceae family, which Prevotella genus belongs to, was obviously upregulated in Atf3−/− mice (Fig. 2C, 2D). Evaluation of the representative species of Prevotella genus showed that P. copri was clearly increased in Atf3−/− mice when compared with WT control (Fig. 2E, 2F).

We next sought to determine whether the dysbiosis observed in Atf3−/− mice was caused by the defective TFH/IgA axis, intestinal microbiota from CD4creAtf3fl/fl mice, and Atf3fl/fl littermates were evaluated. Consistently, flow cytometric analysis showed that the proportion of IgA-bound bacteria was significantly reduced in CD4creAtf3fl/fl mice when compared with Atf3fl/fl control (Supplemental Fig. 4A). Further 16S rRNA sequencing of fecal DNA confirmed a clear distinction in microbiota composition between CD4creAtf3fl/fl and Atf3fl/fl mice (Supplemental Fig. 4B–F). Prevotella taxa were consistently increased in CD4creAtf3fl/fl mice (Supplemental Fig. 4F). Further qPCR and DNA electrophoresis showed that Prevotella genus (Fig. 2G, 2H), Prevotellaceae family (Fig. 2I, 2J), and P. copri species (Fig. 2K, 2L), were significantly elevated in feces from CD4creAtf3fl/fl mice as compared with Atf3fl/fl littermates. Collectively, these observations reveal a critical role of T cells in the intestinal dysbiosis observed in ATF3-deficient mice.

To determine the causal relationship between the impaired TFH/IgA axis and intestinal dysbiosis in ATF3-deficient mice, adoptive transfer of TFH cells or IgA+ B cells was performed as described previously (21). TFH cells or IgA+ B cells from PPs of CD45.1+-congenic mice were adoptively transferred into CD4creAtf3fl/fl mice, followed by DSS treatment. Fecal samples were collected for qPCR and DNA electrophoresis analysis. Results showed that the abundance of Prevotella taxa was dramatically decreased after adoptive transfer of either TFH cells (Fig. 3A, 3B) or IgA+ B cells (Fig. 3C, 3D) into CD4creAtf3fl/fl mice. These observations indicate that the defective TFH-IgA axis contributes to the intestinal dysbiosis in the absence of ATF3.

To investigate whether the dysbiosis contributes to the aggravated colitis observed in ATF3-deficient mice, WT and Atf3−/− mice were administered with a broad spectrum of antibiotics (including AVNM) in drinking water for 4 wk, followed by exposure to 2.5% DSS (w/v) (Fig. 4A). As expected, total bacteria in gut were apparently reduced after 4 wk of AVNM treatment in both WT and Atf3−/− mice (Fig. 4B). Depletion of microbiota significantly alleviated the disease severity of colitis in Atf3−/− mice, which were comparable to AVNM-treated WT littermates. The disease symptoms of colitis were represented by body weight loss, DAI, and shortened colon length (Fig. 4C–F). The alleviation of colitis was further confirmed by histological staining of colon tissues and the quantitated colitis score (Fig. 4G, 4H). Collectively, these data indicate that gut microbiota from Atf3−/− mice aggravated colitis.

To further confirm the causal relationship between the gut dysbiosis and aggravated colitis in Atf3−/− mice, fecal transplantation experiments were performed. WT and Atf3−/− mice were initially treated with AVNM to deplete gut microbiota, followed by fecal transplantation from either WT or Atf3−/− mice every 3 d for 3 wk. Mice were then subjected to DSS challenge (Fig. 5A). The abundance of total microbiota was examined to confirm the efficiency of AVNM treatment and fecal transplantation (Fig. 5B). Evaluation of colitis symptoms showed that the recipients receiving microbiota from Atf3−/− mice displayed significantly higher susceptibility to colitis when compared with those received microbiota from WT controls (Fig. 5C–H). Moreover, transplantation of microbiota from WT mice clearly alleviated the colitis severity in Atf3−/− recipients (Fig. 5C–H). These observations indicate that gut microbiota from Atf3−/− mice enhance the susceptibility of colitis in mice.

The colitogenic effect of microbiota from Atf3−/− mice on colitis was further alternatively verified in T cell–mediated colitis model. Rag1−/− mice were administered with antibiotics for 2 wk, followed by fecal transplantation from either WT or Atf3−/− mice for 8 wk. Two weeks after the initiation of fecal transplantation, mice were i.p. transferred with naive CD4+ T cells (CD4+CD25CD45RBhi) (Fig. 6A). The total abundance of gut microbiota was evaluated to confirm the efficiency of AVNM treatment and fecal transplantation (Fig. 6B). Consistently, it was found that Rag1−/− mice colonized with Atf3−/− feces displayed much severer symptoms of colitis, including sharper body weight loss (Fig. 6C), shorter colon length (Fig. 6D, 6E), as well as increased colonic inflammation severity indicated by histological staining and colon histology scores (Fig. 6F, 6G). Taken together, these observations suggest that the effect of microbiota from Atf3−/− mice on colitis is a common finding.

IgA is the most abundant Ig in the gut. The interaction between IgA and commensal bacteria plays a critical role in the maintenance of gut mucosal homeostasis. Recently, several molecules were reported to mediate the interaction between IgA and microbiota in gut, including the inhibitory coreceptor PD-1 (38), innate adaptor MyD88 (28), and the ATP-gated ionotropic P2 × 7 receptor (39). Furthermore, the interaction between selected IgA and the diversified microbiota facilitated the expansion of Foxp3+ T cells, which, in turn, induced GCs reaction and IgA production by forming a symbiotic regulatory loop in the gut (40). In this study, we demonstrated that stress-inducible transcription factor ATF3 represents another molecule mediating the cross-talk between TFH-dependent IgA production and microbiota in gut.

It was reported that IgA-bound bacteria were prone to be colitogenic, and therefore promoting the development of colitis in both adults and infants (36, 41). In this study, a dramatic reduction in the proportion of IgA-bound bacterial was observed in both Atf3−/− and CD4creAtf3fl/fl mice when compared with their littermate controls. Further 16S rRNA sequencing analysis failed to show significant differences in the composition of IgA-bound microbiota between ATF3-deficient and ATF3-sufficient mice (data not shown). It is possible that the defective IgA production in ATF3-deficient mice leads to less pathogenic bacteria bound by IgA, and therefore facilitating their invasion into the gut mucosal barrier and eventually aggravating colitis.

The most enriched bacteria in gut were two phyla: Bacteroidetes and Firmicutes (42). Prevotella belongs to the Prevotellaceae family and represents one genus microbe of Bacteroidetes. Prevotella is more prevalent in non-Westernized population who prefer plant-rich diet (43). However, there were several reports indicating that Prevotella genus and its species P. copri participate in the pathogenesis of chronic inflammatory disorders such as colitis (44), rheumatoid arthritis (26), osteomyelitis (45), and ankylosing spondylitis (46). Animals colonized with P. copri showed higher susceptibility to DSS-induced colitis (26). Mice deficient in IL-17F were resistant to colitis and displayed reduced abundance in Prevotella (47). These investigations suggested that Prevotella may promote inflammation in gut. In this study, it was found that Prevotella genus, especially P. copri species, was remarkably elevated in ATF3-deficient mice when compared with littermate controls. This was consistent with the aggravated colitis in ATF3-deficient mice, indicating that the elevation of P. copri may drive murine colitis in the absence of ATF3. Depletion of P. copri in ATF3-deficient mice may be helpful to clarify this possibility.

Although the deficiency of ATF3 caused aggravated colitis in mice, there were no available reports in public databases regarding ATF3 mutations in patients suffering intestinal inflammation, including The Human Gene Mutation Database, Online Mendelian Inheritance in Man, and ClinVar of National Center for Biotechnology Information. We previously showed that ATF3 expression in colonic tissues from ulcerative colitis patients was negatively correlated with the severity of ulcerative colitis (21). The clinical significance of ATF3 in colitis and dysbiosis of gut microbiota, however, deserves further investigation.

In conclusion, this study reveals ATF3 as an important mediator of IgA-microbiota interaction in gut, which acts as a protective mechanism underlying intestinal homeostasis.

This work was supported by the following grants to J.Z.: National Natural Science Foundation of China 81925018 and 81771665, the Start-Up Fund for High-Level Talents of Tianjin Medical University, Natural Science Foundation of Guangdong Province 2017B030311014, and Science and Technology Program of Guangzhou 201605122045238. This work was also supported by the following grants to Y.C.: Guangdong Basic Research Foundation 2019A1515010029 and China Postdoctoral Science Foundation 2019M650224, and by the following grants to H.W.: National Key Research and Development Program of China 2016YFA0502202 and Strategic Priority Research Program of the Chinese Academy of Sciences XDB29030103.

The online version of this article contains supplemental material.

Abbreviations used in this article:

ATF3

activating transcription factor 3

AVNM

mixture of 1 g/l ampicillin, 0.5 g/l vancomycin, 1 g/l neomycin sulfate, and 1 g/l metronidazole

DAI

disease activity index

DSS

dextran sulfate sodium

GC

germinal center

OTU

operational taxonomic unit

PP

Peyer patch

qPCR

quantitative PCR

sIgA

secretory Ig A

TFH

follicular helper T

WT

wild-type.

1
Stephens
,
W. Z.
,
J. L.
Round
.
2014
.
IgA targets the troublemakers.
Cell Host Microbe
16
:
265
267
.
2
Kato
,
L. M.
,
S.
Kawamoto
,
M.
Maruya
,
S.
Fagarasan
.
2014
.
Gut TFH and IgA: key players for regulation of bacterial communities and immune homeostasis.
Immunol. Cell Biol.
92
:
49
56
.
3
Bunker
,
J. J.
,
A.
Bendelac
.
2018
.
IgA responses to microbiota.
Immunity
49
:
211
224
.
4
Bunker
,
J. J.
,
T. M.
Flynn
,
J. C.
Koval
,
D. G.
Shaw
,
M.
Meisel
,
B. D.
McDonald
,
I. E.
Ishizuka
,
A. L.
Dent
,
P. C.
Wilson
,
B.
Jabri
, et al
.
2015
.
Innate and adaptive humoral responses coat distinct commensal bacteria with immunoglobulin A.
Immunity
43
:
541
553
.
5
Okai
,
S.
,
F.
Usui
,
S.
Yokota
,
Y.
Hori-I
,
M.
Hasegawa
,
T.
Nakamura
,
M.
Kurosawa
,
S.
Okada
,
K.
Yamamoto
,
E.
Nishiyama
, et al
.
2016
.
High-affinity monoclonal IgA regulates gut microbiota and prevents colitis in mice.
Nat. Microbiol.
1
:
16103
.
6
Honda
,
K.
2015
.
TFH-IgA responses keep microbiota in check.
Cell Host Microbe
17
:
144
146
.
7
Bäckhed
,
F.
,
R. E.
Ley
,
J. L.
Sonnenburg
,
D. A.
Peterson
,
J. I.
Gordon
.
2005
.
Host-bacterial mutualism in the human intestine.
Science
307
:
1915
1920
.
8
Brestoff
,
J. R.
,
D.
Artis
.
2013
.
Commensal bacteria at the interface of host metabolism and the immune system.
Nat. Immunol.
14
:
676
684
.
9
Lloyd-Price
,
J.
,
C.
Arze
,
A. N.
Ananthakrishnan
,
M.
Schirmer
,
J.
Avila-Pacheco
,
T. W.
Poon
,
E.
Andrews
,
N. J.
Ajami
,
K. S.
Bonham
,
C. J.
Brislawn
, et al
IBDMDB Investigators
.
2019
.
Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases.
Nature
569
:
655
662
.
10
Cullender
,
T. C.
,
B.
Chassaing
,
A.
Janzon
,
K.
Kumar
,
C. E.
Muller
,
J. J.
Werner
,
L. T.
Angenent
,
M. E.
Bell
,
A. G.
Hay
,
D. A.
Peterson
, et al
.
2013
.
Innate and adaptive immunity interact to quench microbiome flagellar motility in the gut.
Cell Host Microbe
14
:
571
581
.
11
Pabst
,
O.
,
V.
Cerovic
,
M.
Hornef
.
2016
.
Secretory IgA in the coordination of establishment and maintenance of the microbiota.
Trends Immunol.
37
:
287
296
.
12
Yin
,
X.
,
J. W.
Dewille
,
T.
Hai
.
2008
.
A potential dichotomous role of ATF3, an adaptive-response gene, in cancer development.
Oncogene
27
:
2118
2127
.
13
Thompson
,
M. R.
,
D.
Xu
,
B. R. G.
Williams
.
2009
.
ATF3 transcription factor and its emerging roles in immunity and cancer.
J. Mol. Med. (Berl.)
87
:
1053
1060
.
14
Gilchrist
,
M.
,
V.
Thorsson
,
B.
Li
,
A. G.
Rust
,
M.
Korb
,
J. C.
Roach
,
K.
Kennedy
,
T.
Hai
,
H.
Bolouri
,
A.
Aderem
.
2006
.
Systems biology approaches identify ATF3 as a negative regulator of toll-like receptor 4. [Published erratum appears in 2008 Nature 451: 1022.]
Nature
441
:
173
178
.
15
Khuu
,
C. H.
,
R. M.
Barrozo
,
T.
Hai
,
S. L.
Weinstein
.
2007
.
Activating transcription factor 3 (ATF3) represses the expression of CCL4 in murine macrophages.
Mol. Immunol.
44
:
1598
1605
.
16
Gilchrist
,
M.
,
W. R.
Henderson
Jr.
,
A. E.
Clark
,
R. M.
Simmons
,
X.
Ye
,
K. D.
Smith
,
A.
Aderem
.
2008
.
Activating transcription factor 3 is a negative regulator of allergic pulmonary inflammation.
J. Exp. Med.
205
:
2349
2357
.
17
Hoetzenecker
,
W.
,
B.
Echtenacher
,
E.
Guenova
,
K.
Hoetzenecker
,
F.
Woelbing
,
J.
Brück
,
A.
Teske
,
N.
Valtcheva
,
K.
Fuchs
,
M.
Kneilling
, et al
.
2011
.
ROS-induced ATF3 causes susceptibility to secondary infections during sepsis-associated immunosuppression.
Nat. Med.
18
:
128
134
.
18
Smith
,
C. K.
,
N. L.
Seto
,
A.
Vivekanandan-Giri
,
W.
Yuan
,
M. P.
Playford
,
Z.
Manna
,
S. A.
Hasni
,
R.
Kuai
,
N. N.
Mehta
,
A.
Schwendeman
, et al
.
2017
.
Lupus high-density lipoprotein induces proinflammatory responses in macrophages by binding lectin-like oxidised low-density lipoprotein receptor 1 and failing to promote activating transcription factor 3 activity.
Ann. Rheum. Dis.
76
:
602
611
.
19
Bambouskova
,
M.
,
L.
Gorvel
,
V.
Lampropoulou
,
A.
Sergushichev
,
E.
Loginicheva
,
K.
Johnson
,
D.
Korenfeld
,
M. E.
Mathyer
,
H.
Kim
,
L. H.
Huang
, et al
.
2018
.
Electrophilic properties of itaconate and derivatives regulate the IκBζ-ATF3 inflammatory axis.
Nature
556
:
501
504
.
20
Glal
,
D.
,
J. N.
Sudhakar
,
H. H.
Lu
,
M. C.
Liu
,
H. Y.
Chiang
,
Y. C.
Liu
,
C. F.
Cheng
,
J. W.
Shui
.
2018
.
ATF3 sustains IL-22-induced STAT3 phosphorylation to maintain mucosal immunity through inhibiting phosphatases.
Front. Immunol.
9
:
2522
.
21
Cao
,
Y.
,
Q.
Yang
,
H.
Deng
,
J.
Tang
,
J.
Hu
,
H.
Liu
,
M.
Zhi
,
L.
Ye
,
B.
Zou
,
Y.
Liu
, et al
.
2019
.
Transcriptional factor ATF3 protects against colitis by regulating follicular helper T cells in Peyer’s patches.
Proc. Natl. Acad. Sci. USA
116
:
6286
6291
.
22
Wirtz
,
S.
,
V.
Popp
,
M.
Kindermann
,
K.
Gerlach
,
B.
Weigmann
,
S.
Fichtner-Feigl
,
M. F.
Neurath
.
2017
.
Chemically induced mouse models of acute and chronic intestinal inflammation.
Nat. Protoc.
12
:
1295
1309
.
23
Llewellyn
,
S. R.
,
G. J.
Britton
,
E. J.
Contijoch
,
O. H.
Vennaro
,
A.
Mortha
,
J. F.
Colombel
,
A.
Grinspan
,
J. C.
Clemente
,
M.
Merad
,
J. J.
Faith
.
2018
.
Interactions between diet and the intestinal microbiota alter intestinal permeability and colitis severity in mice.
Gastroenterology
154
:
1037
1046.e2
.
24
Rakoff-Nahoum
,
S.
,
J.
Paglino
,
F.
Eslami-Varzaneh
,
S.
Edberg
,
R.
Medzhitov
.
2004
.
Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis.
Cell
118
:
229
241
.
25
Horai
,
R.
,
C. R.
Zárate-Bladés
,
P.
Dillenburg-Pilla
,
J.
Chen
,
J. L.
Kielczewski
,
P. B.
Silver
,
Y.
Jittayasothorn
,
C. C.
Chan
,
H.
Yamane
,
K.
Honda
,
R. R.
Caspi
.
2015
.
Microbiota-dependent activation of an autoreactive T cell receptor provokes autoimmunity in an immunologically privileged site.
Immunity
43
:
343
353
.
26
Scher
,
J. U.
,
A.
Sczesnak
,
R. S.
Longman
,
N.
Segata
,
C.
Ubeda
,
C.
Bielski
,
T.
Rostron
,
V.
Cerundolo
,
E. G.
Pamer
,
S. B.
Abramson
, et al
.
2013
.
Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis.
eLife
2
: e01202.
27
Lamas
,
B.
,
M. L.
Richard
,
V.
Leducq
,
H. P.
Pham
,
M. L.
Michel
,
G.
Da Costa
,
C.
Bridonneau
,
S.
Jegou
,
T. W.
Hoffmann
,
J. M.
Natividad
, et al
.
2016
.
CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands.
Nat. Med.
22
:
598
605
.
28
Kubinak
,
J. L.
,
C.
Petersen
,
W. Z.
Stephens
,
R.
Soto
,
E.
Bake
,
R. M.
O’Connell
,
J. L.
Round
.
2015
.
MyD88 signaling in T cells directs IgA-mediated control of the microbiota to promote health.
Cell Host Microbe
17
:
153
163
.
29
Findley
,
K.
,
J.
Oh
,
J.
Yang
,
S.
Conlan
,
C.
Deming
,
J. A.
Meyer
,
D.
Schoenfeld
,
E.
Nomicos
,
M.
Park
,
H. H.
Kong
,
J. A.
Segre
;
NIH Intramural Sequencing Center Comparative Sequencing Program
.
2013
.
Topographic diversity of fungal and bacterial communities in human skin.
Nature
498
:
367
370
.
30
Tang
,
C.
,
T.
Kamiya
,
Y.
Liu
,
M.
Kadoki
,
S.
Kakuta
,
K.
Oshima
,
M.
Hattori
,
K.
Takeshita
,
T.
Kanai
,
S.
Saijo
, et al
.
2015
.
Inhibition of dectin-1 signaling ameliorates colitis by inducing Lactobacillus-mediated regulatory T cell expansion in the intestine.
Cell Host Microbe
18
:
183
197
.
31
Bergström
,
A.
,
T. R.
Licht
,
A.
Wilcks
,
J. B.
Andersen
,
L. R.
Schmidt
,
H. A.
Grønlund
,
L. K.
Vigsnaes
,
K. F.
Michaelsen
,
M. I.
Bahl
.
2012
.
Introducing GUt low-density array (GULDA): a validated approach for qPCR-based intestinal microbial community analysis.
FEMS Microbiol. Lett.
337
:
38
47
.
32
Elinav
,
E.
,
T.
Strowig
,
A. L.
Kau
,
J.
Henao-Mejia
,
C. A.
Thaiss
,
C. J.
Booth
,
D. R.
Peaper
,
J.
Bertin
,
S. C.
Eisenbarth
,
J. I.
Gordon
,
R. A.
Flavell
.
2011
.
NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis.
Cell
145
:
745
757
.
33
Dziarski
,
R.
,
S. Y.
Park
,
D. R.
Kashyap
,
S. E.
Dowd
,
D.
Gupta
.
2016
.
Pglyrp-regulated gut microflora Prevotella falsenii, parabacteroides distasonis and Bacteroides eggerthii enhance and Alistipes finegoldii attenuates colitis in mice.
PLoS One
11
: e0146162.
34
Braga
,
R. R.
,
M. A.
Carvalho
,
O.
Bruña-Romero
,
R. E.
Teixeira
,
J. E.
Costa
,
E. N.
Mendes
,
L. M.
Farias
,
P. P.
Magalhães
.
2010
.
Quantification of five putative periodontal pathogens in female patients with and without chronic periodontitis by real-time polymerase chain reaction.
Anaerobe
16
:
234
239
.
35
Yu
,
C.
,
B.
Zhou
,
X.
Xia
,
S.
Chen
,
Y.
Deng
,
Y.
Wang
,
L.
Wu
,
Y.
Tian
,
B.
Zhao
,
H.
Xu
,
L.
Yang
.
2019
.
Prevotella copri is associated with carboplatin-induced gut toxicity.
Cell Death Dis.
10
:
714
.
36
Gopalakrishna
,
K. P.
,
B. R.
Macadangdang
,
M. B.
Rogers
,
J. T.
Tometich
,
B. A.
Firek
,
R.
Baker
,
J.
Ji
,
A. H. P.
Burr
,
C.
Ma
,
M.
Good
, et al
.
2019
.
Maternal IgA protects against the development of necrotizing enterocolitis in preterm infants.
Nat. Med.
25
:
1110
1115
.
37
Berry
,
D.
,
C.
Schwab
,
G.
Milinovich
,
J.
Reichert
,
K.
Ben Mahfoudh
,
T.
Decker
,
M.
Engel
,
B.
Hai
,
E.
Hainzl
,
S.
Heider
, et al
.
2012
.
Phylotype-level 16S rRNA analysis reveals new bacterial indicators of health state in acute murine colitis.
ISME J.
6
:
2091
2106
.
38
Kawamoto
,
S.
,
T. H.
Tran
,
M.
Maruya
,
K.
Suzuki
,
Y.
Doi
,
Y.
Tsutsui
,
L. M.
Kato
,
S.
Fagarasan
.
2012
.
The inhibitory receptor PD-1 regulates IgA selection and bacterial composition in the gut.
Science
336
:
485
489
.
39
Perruzza
,
L.
,
G.
Gargari
,
M.
Proietti
,
B.
Fosso
,
A. M.
D’Erchia
,
C. E.
Faliti
,
T.
Rezzonico-Jost
,
D.
Scribano
,
L.
Mauri
,
D.
Colombo
, et al
.
2017
.
T follicular helper cells promote a beneficial gut ecosystem for host metabolic homeostasis by sensing microbiota-derived extracellular ATP.
Cell Rep.
18
:
2566
2575
.
40
Kawamoto
,
S.
,
M.
Maruya
,
L. M.
Kato
,
W.
Suda
,
K.
Atarashi
,
Y.
Doi
,
Y.
Tsutsui
,
H.
Qin
,
K.
Honda
,
T.
Okada
, et al
.
2014
.
Foxp3(+) T cells regulate immunoglobulin a selection and facilitate diversification of bacterial species responsible for immune homeostasis.
Immunity
41
:
152
165
.
41
Palm
,
N. W.
,
M. R.
de Zoete
,
T. W.
Cullen
,
N. A.
Barry
,
J.
Stefanowski
,
L.
Hao
,
P. H.
Degnan
,
J.
Hu
,
I.
Peter
,
W.
Zhang
, et al
.
2014
.
Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel disease.
Cell
158
:
1000
1010
.
42
Ley
,
R. E.
2016
.
Gut microbiota in 2015: Prevotella in the gut: choose carefully.
Nat. Rev. Gastroenterol. Hepatol.
13
:
69
70
.
43
Tett
,
A.
,
K. D.
Huang
,
F.
Asnicar
,
H.
Fehlner-Peach
,
E.
Pasolli
,
N.
Karcher
,
F.
Armanini
,
P.
Manghi
,
K.
Bonham
,
M.
Zolfo
, et al
.
2019
.
The Prevotella copri complex comprises four distinct clades underrepresented in westernized populations.
Cell Host Microbe
26
:
666
679.e7
.
44
Lucke
,
K.
,
S.
Miehlke
,
E.
Jacobs
,
M.
Schuppler
.
2006
.
Prevalence of Bacteroides and Prevotella spp. in ulcerative colitis.
J. Med. Microbiol.
55
:
617
624
.
45
Lukens
,
J. R.
,
P.
Gurung
,
P.
Vogel
,
G. R.
Johnson
,
R. A.
Carter
,
D. J.
McGoldrick
,
S. R.
Bandi
,
C. R.
Calabrese
,
L.
Vande Walle
,
M.
Lamkanfi
,
T. D.
Kanneganti
.
2014
.
Dietary modulation of the microbiome affects autoinflammatory disease.
Nature
516
:
246
249
.
46
Wen
,
C.
,
Z.
Zheng
,
T.
Shao
,
L.
Liu
,
Z.
Xie
,
E.
Le Chatelier
,
Z.
He
,
W.
Zhong
,
Y.
Fan
,
L.
Zhang
, et al
.
2017
.
Quantitative metagenomics reveals unique gut microbiome biomarkers in ankylosing spondylitis. [Published erratum appears in 2017 Genome Biol. 18: 214.]
Genome Biol.
18
:
142
.
47
Tang
,
C.
,
S.
Kakuta
,
K.
Shimizu
,
M.
Kadoki
,
T.
Kamiya
,
T.
Shimazu
,
S.
Kubo
,
S.
Saijo
,
H.
Ishigame
,
S.
Nakae
,
Y.
Iwakura
.
2018
.
Suppression of IL-17F, but not of IL-17A, provides protection against colitis by inducing Treg cells through modification of the intestinal microbiota.
Nat. Immunol.
19
:
755
765
.

The authors have no financial conflicts of interest.

Supplementary data