Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

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

Https

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

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2016 Mar;9(2):336-51.
doi: 10.1038/mi.2015.64. Epub 2015 Jul 15.

Intestinal CD103(+)CD11b(-) dendritic cells restrain colitis via IFN-γ-induced anti-inflammatory response in epithelial cells

Affiliations

Intestinal CD103(+)CD11b(-) dendritic cells restrain colitis via IFN-γ-induced anti-inflammatory response in epithelial cells

A R B M Muzaki et al. Mucosal Immunol. 2016 Mar.

Abstract

A crosstalk between commensals, gut immune cells, and colonic epithelia is required for a proper function of intestinal mucosal barrier. Here we investigated the importance of two distinct intestinal dendritic cell (DC) subsets in controlling intestinal inflammation. We show that Clec9A-diphtheria toxin receptor (DTR) mice after depletion of CD103(+)CD11b(-) DCs developed severe, low-dose dextran sodium sulfate (DSS)-induced colitis, whereas the lack of CD103(+)CD11b(+) DCs in Clec4a4-DTR mice did not exacerbate intestinal inflammation. The CD103(+)CD11b(-) DC subset has gained a functional specialization that able them to repress inflammation via several epithelial interferon-γ (IFN-γ)-induced proteins. Among others, we identified that epithelial IDO1 and interleukin-18-binding protein (IL-18bp) were strongly modulated by CD103(+)CD11b(-) DCs. Through its preferential property to express IL-12 and IL-15, this particular DC subset can induce lymphocytes in colonic lamina propria and in epithelia to secrete IFN-γ that then can trigger a reversible early anti-inflammatory response in intestinal epithelial cells.

PubMed Disclaimer

Figures

Figure 1
Figure 1
Transcriptome of colon dendritic cell (DC) subsets. (a) Sorting strategy for colon DC isolation. Large intestines obtained from 8 mice, either control (steady state (SS)) or dextran sodium sulfate (DSS) treated (day 4 DSS), were pooled and lamina propria (LP) cells were isolated. Procedure was repeated three times independently. Following CD11chighMHCII+ DC subsets were sorted and analyzed in a gene expression microarray: (1) CD103+CD11b, (2) CD103+CD11b+, and (3) CD103CD11b+. (b) Transcript heat map of the ∼640 genes that are at least twofold differentially expressed in one comparison (red: upregulated; green: downregulated). Clustering was performed using Pearson's correlation and complete linkage. Heat map was z-score normalized by row. (c) XY plot of the first two components of a principal component analysis (PCA) of all six groups (SS 1–3 and DSS 1–3). (d) Heat map showing differential expression of selected genes involved in DC development and function; heat map was generated as described in b.
Figure 2
Figure 2
Distinct intestinal myeloid cells are ablated in Clec9A- and Clec4a4-–diphtheria toxin receptor (DTR) mice. Colon cells were obtained from DT-treated CX3CR1GFP wild-type (WT) controls, CX3CR1GFP/Clec9A-DTR, and CX3CR1GFP/Clec4a4-DTR mice. (a) Colon lamina propria (LP) cells were analyzed for CD103 and CD11b expression by gating on CD11chighMHC II+ cells (gate 1) and for CX3CR1 and CD64 expression by gating CD11cintMHC II+ cells (gate 2). (b) Mesenteric lymph nodes (MLNs) were obtained from the same mice and analyzed for CD103 and CD11b expression by gating on CD11cintMHCII++ migratory dendritic cells (DCs; gate 3) and classical lymphoid CD11chighMHC II+ DCs (gate 4). Representative dot plots of colons and MLNs isolated from three different mice are shown. Indicated numbers show the percentage of each gated cell subset.
Figure 3
Figure 3
Differential localization of distinct colon lamina propria (LP) dendritic cell (DC) subsets and CX3CR1+ macrophages at steady state and during dextran sodium sulfate (DSS)-mediated colitis. (a) Representative confocal images of Clec9A+ and Clec4a4+ DC subsets in the colonic innate lymphoid follicles (ILFs). Sections were stained with phycoerythrin (PE)-labeled anti-Clec9A antibody (red), allophycocyanin (APC)-labeled anti-Clec4a4 (blue), and fluorescein isothiocyanate (FITC)-labeled anti-CD11c (green). Original magnification × 20. Bars=50 μm. (b) Representative confocal images of Clec9A+ and Clec4a4+ DC subsets in the colonic LP under steady-state conditions and 4 days (4 d) after DSS treatment. Cryosections were stained as above in (a). Original magnification × 20. Bars=50 μm. (c) Representative confocal images of CX3CR1GFP+ cells (green) in the colonic LP under steady-state (SS) conditions and 4 days after DSS treatment. Sections were stained with 4',6-diamidino-2-phenylindole (DAPI; blue, nuclei). Original magnification × 20. Bars=45 μm. (d) Flow cytometry of colon LP cells of CX3CR1GFP mice analyzed at steady state and after 4 days of DSS. CD11c/major histocompatibility complex (MHC class II)-positive cells were analyzed for CD64/CX3CR1GFP. Indicated numbers show the percentage of each gated cell subset. (e) Percentage of differentially expressing CX3CR1 populations in the colon during steady state and upon 4 days of DSS treatment. N=5 mice, Student's t-test ***P>0.001. (f) DC ablation profiles during DSS treatment. LP cells were collected at day 4 from the distal part of colon of DT-treated wild-type (WT), Clec9A–diphtheria toxin receptor (DTR), and Clec4a4-DTR (n=3 each group). Representative fluorescence-activated cell sorting (FACS) dot plots showing the staining profile of CD11c and MHC class II as well as CD103 and CD11b.
Figure 4
Figure 4
Ablation of CD103+CD11b cells enhances susceptibility to dextran sodium sulfate (DSS)-induced colitis. Wild-type (WT), Clec9A–diphtheria toxin receptor (DTR), and Clec4a4-DTR mice were injected with 20 ng g−1 DT following the schedule described in Methods. Then, 2% DSS was supplied at day 0 ad libidum in the drinking water for 7 consecutive days followed by drinking water at day 8. (a) Body weight was monitored daily over a period of 15 days. Black circles: DT-treated WT control; black triangles: DT-treated Clec9A-DTR; black squares: DT-treated Clec4a4-DTR. Each group: n=8. Values represent the mean±s.d. Three independent experiments were performed with the same numbers of animals. (b) Measurement of colon length at day 8 (cm) of control WT mice (white bar) and DSS-treated DT-injected WT (black bar), Clec4a4-DTR (striped bar), and Clec9A-DTR (gray bar) mice. Each group: n=8. Values represent the mean±s.d. (c) Fecal samples of DT-injected WT controls (black circles), Clec4a4-DTR (black squares), and Clec9A-DTR (black triangles) mice were collected at day 8 upon DSS treatment and scored for blood content. Each group: n>9 mice. Student's t-test significance: ***P>0.001, NS, not significant. (d, e) Increased influx of inflammatory myeloid cells in the absence of Clec9A+CD103+CD11b dendritic cell (DC) subset at early stages of inflammation (day 4 (d4)). Colon of DT-injected WT, Clec4a4-DTR, and Clec9A-DTR mice were analyzed at day 0 and 4 days upon start of DSS treatment. Representative dot plots of three mice indicating CD11bLy6G+ and CD11bLy6C+ cells (d) and bar chart of 3–4 mice±s.d. Student's t-test significance: **P>0.005. (f) Intestinal permeability as determined by quantifying the amount of fluorescein isothiocyanate (FITC)–dextran levels (μg ml−1) in the serum after its oral gavage. DT-injected WT, Clec9A-DTR, or Clec4a4-DTR mice were tested at days 4 (filled symbols) and 10 (empty symbols) from the beginning of DSS treatment. For each group, 7–9 mice were analyzed. Student t-test significance: *P>0.01; **P>0.005. (g) Clec9A DTR mice do not survive a 5%, high-dose DSS treatment. WT, Clec9A-DTR, and Clec4a4-DTR mice were treated with 5% DSS for 7 consecutive days followed by drinking water at day 8. Body weight was monitored daily over a period of 13 days. Mice were killed when body weight was <75% of their original weight. Black circles: DT-treated WT control; black triangles: DT-treated Clec9A-DTR; white circles: DT-treated Clec4a4-DTR. Each group: n=4. Values represent the mean±s.d.
Figure 5
Figure 5
Depletion of CX3CR1high macrophages leads to severe intestinal inflammation. (a) Flow cytometry analysis of different macrophage and dendritic cell (DC) subsets. CX3CR1-GFP-CD169-DTR and CX3CR1-GFP-WT mice were injected with DT (20 ng per g body weight) and analyzed the following day for the ablation profile of different CD11chighMHC IIhigh DCs and CD11cintMHC II high macrophages, respectively. For DC profiling, anti-CD103 and anti-CD11b were used, whereas for macrophage profiling, cells were stained with anti-CD64 and monitored for CX3CR1 GFP expression. (b–e) Ablation of CX3CR1high macrophages enhances susceptibility to dextran sodium sulfate (DSS)-induced colitis. Wild-type (WT) and CD169–diphtheria toxin receptor (DTR) mice were injected with 20 ng g−1 DT following the schedule described in Methods. (b) Body weight was monitored daily over a period of 15 days. Open circles: DT-treated WT control; filled circles: DT-treated CD169-DTR. Each group: n=5. Values represent the mean±s.d. Two independent experiments were performed with the same numbers of animals. (c) Fecal samples of DT-injected WT controls (open circles) and CD169-DTR (filled circles) mice were collected at day 8 upon DSS treatment and scored for blood content. Each group: n>5 mice. Student's t-test significance: ***P>0.001. (d) Measurement of colon length at day 8 (cm) of control WT mice (gray bar) and DSS-treated DT-injected WT (white bar) or CD169 DTR (black bar) mice. Each group: n=5. Values represent the mean±s.d. (e) Intestinal permeability as determined by quantifying the amount of fluorescein isothiocyanate (FITC)–dextran levels (μg ml−1) in the serum after its oral gavage. DT-injected WT (open circles) and CD169-DTR mice (filled circles) were tested at days 4 and 10 from the beginning of DSS treatment. For each group, 5–9 mice were analyzed.
Figure 6
Figure 6
Epithelial expressed interferon-γ (IFN-γ)-inducible genes are strongly affected by ablation of CD103+CD11b dendritic cells (DCs). (a) Heat map showing differential expression of selected genes regulated by IFN-γ of colon intestinal epithelial cells (IECs) obtained from wild-type (WT) untreated mice and dextran sodium sulfate (DSS)-treated day 4 WT and Clec9A–diphtheria toxin receptor (DTR) mice (n=3). (b) Gene validation comparing bulk IECs and CD45+ lymphocyte-depleted IECs obtained from DSS-treated animals. IECs were isolated from the colon as described in Methods and loaded on a Percoll gradient to separate the lymphocytes from the epithelial fraction. RNA and subsequently complementary DNA (cDNA) was prepared and validated for Cd3, Ifn-γ, and a series of Ifn-γ-induced genes, including Ido1 and IL-18bp. One representative sample is shown. (c) Quantitative real-time PCR (qPCR) analysis of Ido1 expression in different intestinal DC subsets and IECs at steady state (SS) and 4 days after DSS treatment. N=3±s.e.m. (d) Indoleamine 2,3 dioxygenase (IDO1) is the major tryptophan-degrading enzyme in the colonocytes. IECs obtained from distal part of the colon of DSS-treated WT mice (day 4) were analyzed for Ido1, Ido2, and Tdo expression by semiquantitative real-time PCR (RT-PCR) analysis. Hprt was used as an endogenous mRNA control. Results are representative of 3–4 pooled colons. (e) Ido1 and IL-18bp expression profile during DSS treatment in IECs. WT mice were treated with 1% DSS over 6 days. Colonocytes were isolated from the distal part of three mice every day and monitored by RT-PCR for Ido1 and IL-18bp mRNA expression. (f) qPCR analysis of IL-18bp expression in IECs at steady state and 4 days after DSS treatment. N=3±s.e.m. (g) RT-PCR analysis of Ido1 and IL-18bp in IECs obtained from pooled colons of DT-injected untreated or DSS-treated (day 4) WT, Clec9A-DTR, and Clec4a4-DTR mice. PCR results are representative of three independent IEC isolations. (h) IDO1 protein expression in IECs pooled from three DSS-treated WT or Clec9A DTR mice (day 4). Representative immunoblots for epithelial IDO1 (45 kDa) and β-tubulin control (50 kDa) are shown. (i) Absence of CX3CR1high macrophages does not affect expression of IDO1 and interleukin-18-binding protein (IL-18bp) in IECs during colitis. RT-PCR analysis of Ido1and IL-18bp in IECs obtained from DT-injected untreated or DSS-treated (day 4) WT and CD169-DTR mice. PCR results are representative of three independent IEC isolations.
Figure 7.
Figure 7.
IFN-γ −/− mice show enhanced susceptibility to dextran sodium sulfate (DSS)-induced colitis. Wild-type (WT) and interferon-γ (IFN-γ)−/− mice were treated as described in Methods. (a) Body weight was monitored daily over a period of 11 days. IFN-γ−/− mice were killed at day 8 because of severe body weight loss (>30%). White circles: CB57/BL6 control; black circles: IFN-γ−/− mice. Each group: n=5. Values represent the mean±s.d. Two independent experiments were performed with the same numbers of animals. (b) Fecal samples of CB57/BL6 control and IFN-γ −/− mice were collected at day 7 upon DSS treatment and scored for blood content. Each group: n>7 mice. Student's t-test significance: ****P>0.0001.
Figure 8
Figure 8
IDO1 and IL-18bp expression is modulated by interferon-γ (IFN-γ). (a) Colonocytes express IFN-γ receptor (IFN-γR). The ex vivo isolated colonocytes and CMT-93 colon epithelial cell line were analyzed by semiquantitative real-time PCR (RT-PCR) analysis for IFN-γ receptor expression. Hprt was used as an endogenous mRNA control. (b) IDO1 and IL-18bp expression is induced by IFN-γ. CMT-93 cells were stimulated overnight with 100 U ml−1 IFN-γ and analyzed for Ido1 and IL-18bp expression by semiquantitative RT-PCR analysis. (c) IFN-γ−/− mice do not upregulate Ido1 and IL-18bp epithelial expression upon dextran sodium sulfate (DSS) treatment. Intestinal epithelial cells (IECs) were collected from untreated or DSS-treated wild-type (WT) and IFN-γ−/− mice and evaluated by semiquantitative RT-PCR. One representative sample from each experimental group of three mice is shown. SS, steady state. (d) Clec9A–diphtheria toxin receptor (DTR) mice have a decreased proportion of IFN-γ-expressing lamina propria (LP) T cells and intraepithelial lymphocytes (IELs). Representative flow cytometry plots of LP and IELs harvested from wild-type (WT) and Clec9A-DTR mice 4 days after DSS treatment and stained for CD4, CD8, and γ/δ T cell receptor (TCR), respectively (representative fluorescence-activated cell sorting (FACS) dot plot, right panel) and stained for intracellular IFN-γ. Quantification of LP CD4+ T cells, LP CD8+ T cells, LP CD4CD8 T-cell fraction, γ/δ+, and CD8+ IELs expressing IFN-γ. N=6–8 mice pooled from 2 independent experiments±s.e.m. Student's t-test significance: *P>0.01, ***P>0.001, NS, not significant. (e) Quantitative real-time PCR (qPCR) analysis of IL-15, IL-12p40, IL-12p35, and IL-13p19 expression in distinct colon dendritic cell (DC) subsets obtained from control WT mice: CD103+CD11b, CD103+CD11b+, and CD103CD11b+. Data are representative of 3 independent experiments with 10 mice pooled in each group.
Figure 9
Figure 9
Immunostimulatory oligonucleotide (ISS-ODN) treatment limits the colitis severity in Clec9A–diphtheria toxin receptor (DTR) mice. DT-injected wild-type (WT) and Clec9A-DTR mice were injected intraperitoneally (i.p.) 10 μg of ISS-ODN at the start of the dextran sodium sulfate (DSS) treatment (2%) and 4 days later. (a) Interferon-γ (IFN-γ) response was measured in the serum collected at day 4. (b) Epithelial Ido1 and IL-18bp expression profile at steady-state or under DSS treatment. Representative samples of three WT and Clec9A-DTR mice are shown. (c) The body weight was monitored daily over a period of 10 days. Black circles: DT-treated WT control; white circles: DT-treated WT control+ISS-ODN; black squares: DT-treated Clec9A-DTR; white squares: DT-treated Clec9A-DTR+ISS-ODN; Each group: n=6 mice from two independent experiments. Values represent the mean±s.d. ND, not detectable.

Similar articles

Cited by

References

    1. Brown, E.M., Sadarangani, M. & Finlay, B.B. The role of the immune system in governing host-microbe interactions in the intestine. Nat. Immunol. 14, 660–667 (2013). - PubMed
    1. Macdonald, T.T. & Monteleone, G. Immunity, inflammation, and allergy in the gut. Science 307, 1920–1925 (2005). - PubMed
    1. Ponda, P.P. & Mayer, L. Mucosal epithelium in health and disease. Curr. Mol. Med. 5, 549–556 (2005). - PubMed
    1. Schmitz, H. et al. Altered tight junction structure contributes to the impaired epithelial barrier function in ulcerative colitis. Gastroenterology 116, 301–309 (1999). - PubMed
    1. Peeters, M. et al. Clustering of increased small intestinal permeability in families with Crohn's disease. Gastroenterology 113, 802–807 (1997). - PubMed

Publication types

MeSH terms

LinkOut - more resources

-