Dendritic cells (DCs) bear the main responsibility for initiation of adaptive immune responses necessary for antimicrobial immunity. In the small intestine, afferent lymphatics convey Ags and microbial signals to mesenteric lymph nodes (LNs) to induce adaptive immune responses against microbes and food Ags derived from the small intestine. Whether the large intestine is covered by the same lymphatic system or represents its own lymphoid compartment has not been studied until very recently. We identified three small mesenteric LNs, distinct from small intestinal LNs, which drain lymph specifically from the colon, and studied DC responses to the attaching and effacing pathogen Citrobacter rodentium in these. Transcriptional profiling of conventional (CD11chighCD103high) DC and plasmacytoid (plasmacytoid DC Ag-1highB220+CD11cint) DC (pDC) populations during steady-state conditions revealed activity of distinct sets of genes in these two DC subsets, both in small intestinal and colon-draining LNs. C. rodentium activated DC especially in colon-draining LNs, and gene expression changed in pDC more profoundly than in conventional DC. Among the genes most upregulated in pDC were C-type lectin receptor CLEC4E, IL-1Rs (IL-1R1 and -2), proinflammatory cytokines (IL-1a and IL-6), and TLR6. Our results indicate that colon immune surveillance is distinct from that of the small intestine in terms of draining LNs, and identify pDC as active sentinels of colonic inflammation and/or microbial dysbiosis.

Conventional dendritic cells (cDCs) are sentinels of microbe- and danger-associated signals in their local environment and migrate from tissue to draining lymph nodes (LNs) via the lymphatic system. In the gut, the CD103+ DCs are the most predominant population of cDCs. They populate small intestinal lamina propria and migrate to mesenteric LNs (MLN) (1) to induce regulatory and antimicrobial immune responses to food-borne Ags and commensal microbiota (2, 3). The capacity of CD103+ migratory DCs to imprint responding CD4+ T cells with gut-homing receptors CCR9 and integrin α4β7 (4) and to synthetize retinoic acid have placed this DC subset in a central role in inducing tolerance to food-borne Ags and commensal bacteria (5, 6). In parallel with their ability to induce regulatory Foxp3+ regulatory T cells under steady-state conditions, their importance in promoting antimicrobial Th17 helper T cell responses under inflammatory conditions is well documented (7).

Plasmacytoid DCs (pDCs) differ from cDCs in many aspects (reviewed in Ref. 8). They are mainly found in blood and secondary lymphoid tissues (9, 10), but are relatively sparse in peripheral tissues and mostly home in secondary lymphoid tissues directly from circulating blood. Compared to other peripheral tissues, pDC are numerous in intestinal lamina propria (11) of the small intestine. The ability of pDCs to capture Ags and present Ags to T cells appear to be inferior to cDCs, but their ability to sense environmental cues and elaborate mediators of immune response nevertheless points toward an important role in T cell activation and differentiation (reviewed in Ref. 12). pDCs express their unique set of TLR receptors, and, in response to foreign nucleic acids, circulating pDCs produce large quantities of IFN-α. However, in the gut, pDC appear to promote Th17 cells (13) for antimicrobial defense and, during steady-state conditions, to induce regulatory T cells (14). Despite these important aspects, the roles of pDC in intestinal immunity are less well characterized than those of CD103+ DCs (15).

Most of the knowledge on DCs and their roles in intestinal immunity is related to the small intestine. Although colonic mucosa and lamina propria immune cells have been the focus of much research, less attention has been paid to the lymphatic drainage of the colon. Carter and Collins (16) used ink injections to draw a sketch on the lymph drainage from different parts of the intestine already in 1974 and proposed that lymph from colon drains to LNs distinct from the main MLN. More recently, Van den Broeck et al. (17) illustrated lymph drainage from the intestine using modern imaging techniques and confirmed the distinction between lymphatics of the small intestine and colon. Despite this, no data were available to evaluate if immune responses in the small intestine and colon involve anatomically distinct lymphoid structures and cell types. This problem was tackled quite recently by Houston et al. (18), who used photoconvertible reporter techniques and multicolor flow cytometry to document DC accumulation from the colon and small intestine into their respective draining LNs. They identified three novel colon-draining LNs distinct from the main MLN draining small intestine. The unique phenotypes of their CD103+ DC subsets and T cells and the inability of CD103+DCs in these LNs to prime T cells in response to small-intestinal and oral Ags indicated that these LNs and their DC segregate from the immune surveillance of the small intestine (18).

Although the role of colon was long thought to focus mainly on absorption of excess water from luminal contents, recent advances in understanding the importance of colon and its microbiota in host metabolism and maintenance of immune homeostasis has created novel interest in studying the role of colon immune system in health and disease. Under an independent initiative, we set an endeavor to identify the LNs to which lymph drains from the colon to evaluate the roles of cDCs and pDCs in these LNs during microbial infection of the large intestine. Our findings corroborate the existence of three distinct LNs draining the colon, but also identify a role for each one of these in draining the proximal, transverse, or descending section of the colon. We show that microbial challenge of the colon with Citrobacter rodentium infection leads to selective activation of especially pDC in these and identify unique pathogen-sensing and activation pathways in pDC with potential importance in mounting a protective Th1 response (19, 20) to this effacing-attaching pathogen.

C57BL/6NCrl mice used in this study were bred and maintained in Turku University’s central animal facility. All animals were kept under specific pathogen-free housing conditions and under filter lids. Food and water were available ad libitum, and there was a 12-h/12-h light/dark cycle. All experiments were approved by the national board of animal experimentation in Finland under license ESAVI/6082/04.10.07/2014.

Mice were anesthetized using 150 mg/kg ketamine (Ketalar 50 mg/ml; Pfizer) and 10 mg/kg rompun (Rompun vet 20 mg/ml; Bayer) mixture i.p. A small incision was made into the skin, and peritoneal membrane and colon were gently pulled out. FITC-labeled dextran (FITC-Dx; 70 kDa; 80 mg/ml; Sigma-Aldrich) or Alexa Fluor 488–labeled OVA (OVA-488, 1 mg/ml; Life Technologies) was injected into colon wall intraserosally. Separate injections were made into ascending, transverse, and descending parts of the colon. The incision was closed, and mice were kept anesthetized on a warm blanket for 40 min before sacrifice. When analyzing lymph drainage after FITC-Dx, a tissue block including cecum, colon, mesenteric LNs, pancreatic LNs, and pancreas was prepared. The block was glued onto a Petri dish and submerged in cold PBS. FITC signal was analyzed under stereomicroscope (SteREO Lumar V12; Carl Zeiss). To analyze Ag uptake by DCs, LNs were collected and DC populations labeled as described below. Alexa Fluor 488 signal analyzed as mean fluorescence intensity by FACS.

Mice were fed with 1 to 2 × 109 CFU C. rodentium in Luria-Bertani medium or sterile Luria-Bertani (mock) by mouth and sacrificed 1, 3, 5, or 8 d postgavage. Individual LNs were collected and analyzed for DC activation. Shortly, LNs were collected in RPMI 1640 medium and digested with collagenase (10 U/ml, 10 min, 37°C). Collagenase was inactivated by adding 3 ml DMEM with 10% FCS and 1% nonessential amino acid. Digested tissue was pressed through a tight metal mesh and cell suspension filtered through a 77-μm nylon membrane. Unspecific binding was blocked by adding 5% rat serum. In flow cytometer analysis, live single cells were gated and further analyzed as described below. pDCs were identified as pDC Ag-1 (PDCA-1; PE-conjugated clone 927; BioLegend) and B220 (allophycocyanin-conjugated clone RA3-6B2; BioLegend) double-positive cells. Lamina propria–derived DCs were identified as CD11c (PE-conjugated clone N418; BioLegend) and CD103 (allophycocyanin-conjugated clone 2E7; BioLegend) double-positive cells. Both populations were further characterized for expression of CD11c (FITC-conjugated clone N418; BioLegend), CD11b (FITC-conjugated clone M1/70; BD Pharmingen), F4/80 (allophycocyanin-conjugated clone BM8; BioLegend), Siglec-H (PerCP/Cy5.5-conjugated clone 551; BioLegend), Gr1 (Alexa 488–conjugated clone RB6-8C5; eBioscience), and CD8a (FITC-conjugated, catalog number 22150083; ImmunoTools). Activation of these populations were evaluated by abundance of CD80 (FITC-conjugated clone 16-10A1; eBioscience) and CD86 (FITC-conjugated clone GL1; eBioscience). T-lymphocytes were analyzed for IFN-γ and IL-17A production. To analyze cytokine production, harvested cells were stimulated for 4 h in DMEM medium supplemented with 10% FCS, 25 mmol HEPES, 1% nonessential amino acid, and Cell Activation mixture (catalog number 423304; BioLegend). Lymphocytes were stained for CD4 (FITC-conjugated clone RM4-5; BioLegend) and CD8 (allophycocyanin-conjugated clone 53-6.7; BioLegend). Intracellular staining was performed using Transcription Factor buffer set (catalog number 562725; BD Biosciences) according to the manufacturer’s instructions and PE-labeled IFN-γ (clone XMG1.2; BioLegend) or IL-17A (clone TC11-18H10.1; BioLegend) Ab. Stained cells were analyzed with Accuri C6 flow cytometer and Accuri C6 software (BD Biosciences).

To evaluate LN enlargement in response to C. rodentium infection, the size of the largest colon-draining MLN (coMLN) and small intestine–draining MLN (siMLN) was measured at day 3 using an electronic digital caliber. For coMLN, both length and width was measured, and for siMLN, only the thickness of the proximal end, closest to the coMLN, was measured. The volume of coMLN was calculated using the formula for ellipsoid volume (4/3πabc, assuming the ellipsoid shape to be symmetrical, e.g., b = c).

C. rodentium (strain ICC168, from Prof. Fiona Powrie, University of Oxford, Oxford, U.K.) was grown overnight in Luria broth supplemented with nalidixic acid (1 mg/l). Overnight growth was concentrated 10 times by centrifugation. Concentrated bacterial solution was fed to mice by mouth in 200 μl. Bacterial counts were measured by plate assays: serial dilutions were made from feeding solution and plated on Luria agar plates with 50 μg/ml nalidixic acid overnight. Colonies were counted to quantify the fed bacteria.

To analyze infection-derived changes in cytokines and transcription factors, the colon was collected into RNALater (Qiagen, Germantown, MD). Total RNA was extracted with bead-based PowerLyzer RNA isolation kit (MoBio, Carlsbad, CA). cDNA was synthetized with Maxima reverse transcriptase and random hexamer primers (Thermo Fisher, Waltham, MA). For all primer and probe details, see Supplemental Table I. Relative cytokine expression was determined by real-time quantitative PCR (Lightcycler 480; Roche, Basel, Switzerland) using LightCycler 480 Probes Master or LightCycler SYBR Green I Master mastermixes (Roche). All cytokine signals were normalized to β-actin expression and shown as arbitrary relative expression values (1/2[threshold cycle(target) − threshold cycle(reference)]).

Mice were treated with C. rodentium or sterile medium as described above and sacrificed at day 3 postinfection. Three independent experiments were performed, and for each data point, coMLN and siMLN were collected and pooled from four animals. Cell suspensions were made and pDCs stained as described above. To be able to sort both populations from the same tube, CD103+ DCs were stained with FITC-conjugated CD11c and PerCP/Cy5.5-conjugated CD103 (clones N418 and 2E7, respectively; BioLegend). Both populations were sorted simultaneously into the lysis buffer using FACSAria II Cell Sorter (BD Biosciences). The cell yields for CD103+ cells were 14,584–117,457 cells and for pDC cells 1,158–7,020 cells. GenElute LPA (catalog number 5-6575; Sigma-Aldrich) RNA carrier was added (1 μl) into the lysis buffer prior to RNA extraction from pDC and CD103+ DC cells. Total RNA was extracted using RNAeasy Plus Micro kit (catalog number 74034; Qiagen) according to the manufacturer’s instructions. The quantities of the RNA were measured with Nanodrop, and quality was assured with Agilent 2100 Bioanalyzer and Pico RNA Assay (Agilent Technologies) when a sufficient amount of material was available.

Libraries for RNA sequencing were prepared from 10 ng totalRNA with Smart-Seq v4 Ultra Low input RNA kit (catalog numbers 634896 and 634895; Clontech) according to the kit manual. The cDNAs were quantified with ThermoFisher Qubit dsHS assay, and quality was assured with Agilent 2100 Bioanalyzer DNA High Sensitivity assay (Agilent Technologies). From each sample, 150 ng cDNA was amplified with Illumina Nextera XT DNA library kit (catalog number FC-131-1024; Illumina) according to the kit manual (part 15031942 Rev D). Libraries were quantified with ThermoFisher Qubit dsDNA HS assay (Thermo Fisher), and quality was assured with Agilent Bioanalyzer 2100 DNA High Sensitivity Assay (Agilent Technologies). The clonal cluster amplification was carried out with Illumina cBOT System (Illumina). The amplified libraries were run with Illumina HiSeq2500 Next-Generation Sequencing platform (Illumina) in a single lane of a paired-end rapid run with read length of 100 bp. The run generated a total of 1.65 × 108 raw reads passing filter with a Q30% score of 92.5%.

The quality control of raw sequencing reads was performed with FastQC (http://www.bioinformatics.babraham.ac.uk/projects/fastqc/), and adapters and low quality bases were trimmed by Trimmomatic (21). The trimmed reads were then aligned to the mouse reference genome NCBIM37/mm9 (Ensembl release 67) using Tophat2 (22). Htseq-count (23) summarized read counts for each gene. The R/Bioconductor package edgeR (24) was used to identify differentially expressed genes. Genes with a false discovery rate (FDR) <0.05 and an absolute fold change >1.5 were considered significantly differentially expressed. Statistical analyses for quantitative PCR and FACS data were done using GraphPad Prism 5 software (GraphPad, La Jolla, CA). Statistical significance was determined using two-tailed Student t test or one-way ANOVA with Dunnett post hoc test. The p values <0.05 were considered statistically significant.

FITC-Dx was injected subserosally into three different areas of the colon corresponding to the proximal, middle, or distal part of the colon, and the passage of FITC-Dx into afferent lymphatics was visualized using a low-magnification UV stereomicroscope. We identified three individual coMLNs (Fig. 1A, 1B). The ascending colon and right colic flexure drained to a separate LN near cecum adjacent to the MLN. The transverse colon drained to a LN located close to the other end of the siMLNs and between the string of MLNs and the colon, and the descending colon drained to a small LN in the mesenterium in close proximity to the pancreas (Fig. 1). All three LNs have their efferent lymph veins flowing to the same large lymph vein in the mesenterium. As these three LNs appeared to collect afferent lymph specifically from the colon and be distinct from the main part of the MLN draining the small intestine, these three LNs will be collectively referred to as coMLN in distinction from the siMLN.

FIGURE 1.

Anatomical location of coMLN. (A) The largest of the coMLNs (arrow, removed in B) is located near the hepatic flexure of colon adjacent to siMLN. (B) Second coMLN is located close to the opposite end of siMLN (arrow). The third coMLN is usually not visible to the naked eye and is not included in (A) or (B). (C) Schematic representation of colon draining lymphatic system. FITC-Dx was injected subserosally into various sites of the colon (red X). The gut and the mesenterium were removed en bloc and visualized under a stereomicroscope. Several related organs are drawn to help place the fluorescent images into context. (1) Cecum; (2) hepatic flexure and proximal colon; (3) transverse colon; (4) descending colon; (5) siMLN (red); and (6) the pancreas (blue). Lymph from the hepatic flexure and proximal colon is collected to coMLN located at the proximal end of siMLN (D and G). Lymph from the transverse colon is collected to the second coMLN, which is located at the distal end of the siMLN and embedded in pancreas (E and H). Lymph from descending colon is collected to a third coMLN, which is embedded in the pancreas (F and I). Efferent lymph veins from all three coMLNs join into a larger lymph vein (drawn green line). Scale bars, 1 mm. Photographs are representative of six individual experiments.

FIGURE 1.

Anatomical location of coMLN. (A) The largest of the coMLNs (arrow, removed in B) is located near the hepatic flexure of colon adjacent to siMLN. (B) Second coMLN is located close to the opposite end of siMLN (arrow). The third coMLN is usually not visible to the naked eye and is not included in (A) or (B). (C) Schematic representation of colon draining lymphatic system. FITC-Dx was injected subserosally into various sites of the colon (red X). The gut and the mesenterium were removed en bloc and visualized under a stereomicroscope. Several related organs are drawn to help place the fluorescent images into context. (1) Cecum; (2) hepatic flexure and proximal colon; (3) transverse colon; (4) descending colon; (5) siMLN (red); and (6) the pancreas (blue). Lymph from the hepatic flexure and proximal colon is collected to coMLN located at the proximal end of siMLN (D and G). Lymph from the transverse colon is collected to the second coMLN, which is located at the distal end of the siMLN and embedded in pancreas (E and H). Lymph from descending colon is collected to a third coMLN, which is embedded in the pancreas (F and I). Efferent lymph veins from all three coMLNs join into a larger lymph vein (drawn green line). Scale bars, 1 mm. Photographs are representative of six individual experiments.

Close modal

Macroscopic examination of coMLN and siMLN showed that the size of coMLN, but not siMLN, was significantly increased already 3 d after oral inoculation of the pathogen (Fig. 2). In line with earlier reports (2527), cytokines, transcription factors related to activation of macrophages, innate lymphoid cells, T cells, and stress indicators of epithelial cells became upregulated in colon tissue only later, ∼8 d postinfection (Supplemental Fig. 1). This implicates the coMLNs in immune recognition of the pathogen already before prominent inflammation in the gut.

FIGURE 2.

Colon draining LNs increase significantly in size after C. rodentium infection. Colon-draining LN (arrows) became significantly enlarged by 3 d postinfection (AC), whereas no difference was observed in siMLN (C). The thickness of both coMLN and siMLN was measured using electronic calipers. Length of coMLN was also measured and used to calculate the volume of the coMLN assuming an ellipsoid shape. The volume shows significant increase after C. rodentium infection when compared with mock treatment (D). Error shown as SD. **p < 0.01, n = 6.

FIGURE 2.

Colon draining LNs increase significantly in size after C. rodentium infection. Colon-draining LN (arrows) became significantly enlarged by 3 d postinfection (AC), whereas no difference was observed in siMLN (C). The thickness of both coMLN and siMLN was measured using electronic calipers. Length of coMLN was also measured and used to calculate the volume of the coMLN assuming an ellipsoid shape. The volume shows significant increase after C. rodentium infection when compared with mock treatment (D). Error shown as SD. **p < 0.01, n = 6.

Close modal

We studied two different DC populations, cDCs and pDCs in the LNs, which we identified as coMLN. These DC populations were defined by their expression of either PDCA-1 and B220 (pDCs) or CD11c and CD103 (cDCs) (Fig. 3). Additional markers expressed by coMLN-derived pDCs were consistent with known pDC markers (Fig. 3E–G). cDC in coMLN were defined as CD11chigh/CD103+ DC, containing both migratory and resident DC populations (28, 29). Interestingly, although CD11b+ expression was detected on a subpopulation of CD103+ DCs in siMLN (24.01 ± 3.61%), CD11b+CD103+ DCs were rare in coMLN (3.65 ± 3.98%, p = 0.0009). The majority of CD103+ DCs in coMLN expressed CD8a (70.27 ± 12.91%) (Fig. 3H–J). CD11b/CD8a+ cells are the dominant subpopulation of CD103+ DCs in the coMLN (44.10 ± 6.97%, p < 0.0001) when compared with CD11b+/CD8a+ and CD11b+/CD8a fractions.

FIGURE 3.

Gating strategy and population analysis of subsequently studied DC classes. Live single cells were gated and identified either as pDC by PDCA-1/B220 expression or cDCs by CD11c/CD103 expression (AD). pDCs were studied for various population-related markers: CD11c (E), Siglec-H (F), and Ly6C/G (G). CD103+ DCs were analyzed for CD11b (H and I) and CD8a (J) subpopulations. CD11b expression on the CD103+ cDCs was significantly lower and virtually nonexistent in coMLN when compared with siMLN (p = 0.0009, H and I). Black graphs for specific markers, gray for isotype control. Representative histograms are shown with mean percentage ± SD from four individual mice. FSC-A, forward light scatter-area; FSC-H, forward light scatter-height; SSC-A, side scatter-area.

FIGURE 3.

Gating strategy and population analysis of subsequently studied DC classes. Live single cells were gated and identified either as pDC by PDCA-1/B220 expression or cDCs by CD11c/CD103 expression (AD). pDCs were studied for various population-related markers: CD11c (E), Siglec-H (F), and Ly6C/G (G). CD103+ DCs were analyzed for CD11b (H and I) and CD8a (J) subpopulations. CD11b expression on the CD103+ cDCs was significantly lower and virtually nonexistent in coMLN when compared with siMLN (p = 0.0009, H and I). Black graphs for specific markers, gray for isotype control. Representative histograms are shown with mean percentage ± SD from four individual mice. FSC-A, forward light scatter-area; FSC-H, forward light scatter-height; SSC-A, side scatter-area.

Close modal

To confirm lymph drainage from different parts of the colon to the abovementioned LNs, we determined Ag uptake by DCs in coMLN after injection of OVA–Alexa 488 into the colon wall. Both the CD103+ DC and the PDCA-1+/B220+ DC were evaluated. Ag uptake was determined 40 min after injection and therefore mostly represents uptake of soluble material arriving in the LN with the lymph (30). Both DC populations in coMLN displayed Alexa 488 fluorescence (Fig. 4), indicating Ag uptake by DCs in the same LNs as the ones to which afferent lymph vessels from the colon ended (as illustrated by microscopy of the mesenterium, see Fig. 1). Interestingly, pDCs were significantly more efficient in taking up OVA–Alexa 488 than the CD103+ DCs, indicating that they sample Ags directly and more efficiently than CD103+ DCs, which are present in coMLN at time of Ag arrival.

FIGURE 4.

pDCs in coMLN take up soluble Ag draining from the colon. Alexa 488–OVA was injected into the colon wall. LNs were collected, and Alexa 488 signal was analyzed from pDCs (A) and CD103+ DCs (B). (C) The pDCs showed significantly more Ag uptake when compared with CD103+ DCs in coMLN and significantly more in coMLN than in siMLN. Each data point represents one mouse. Error shown as SD. *p < 0.05, **p < 0.01, n = 3. BLN, brachial LN; MFI, mean fluorescence intensity.

FIGURE 4.

pDCs in coMLN take up soluble Ag draining from the colon. Alexa 488–OVA was injected into the colon wall. LNs were collected, and Alexa 488 signal was analyzed from pDCs (A) and CD103+ DCs (B). (C) The pDCs showed significantly more Ag uptake when compared with CD103+ DCs in coMLN and significantly more in coMLN than in siMLN. Each data point represents one mouse. Error shown as SD. *p < 0.05, **p < 0.01, n = 3. BLN, brachial LN; MFI, mean fluorescence intensity.

Close modal

To study the kinetics of DC activation in coMLN, we determined CD80 and CD86 expression on DCs in coMLN and siMLN 1, 3, 5, and 8 d after C. rodentium inoculation. Indeed, both pDC and CD103+ DC upregulated costimulatory ligand expression in response to C. rodentium infection. CD103+ DCs showed upregulation of CD80 (but not CD86) already at day 1 postinfection, and this occurred concomitantly in coMLN and siMLN (Fig. 5). pDCs upregulated both CD80 and CD86, and importantly, this occurred selectively in coMLN by day 3 postinfection. Only later at day 5, CD80 and CD86 upregulation on pDCs was detected also in siMLN (Fig. 5).

FIGURE 5.

pDCs and migratory CD103+ DCs are activated especially in coMLN after C. rodentium infection. DC subsets were identified as pDCs using PDCA-1+ and B220+ expression (A and B) and as cDCs using CD11c+ and CD103+ expression (C and D). CD80 and CD86 expression was analyzed at indicated time points postinfection. See 11Results for interpretation. Error shown as SD. *p < 0.05, **p < 0.01, ***p < 0.001, n = 6. MFI, mean fluorescence intensity.

FIGURE 5.

pDCs and migratory CD103+ DCs are activated especially in coMLN after C. rodentium infection. DC subsets were identified as pDCs using PDCA-1+ and B220+ expression (A and B) and as cDCs using CD11c+ and CD103+ expression (C and D). CD80 and CD86 expression was analyzed at indicated time points postinfection. See 11Results for interpretation. Error shown as SD. *p < 0.05, **p < 0.01, ***p < 0.001, n = 6. MFI, mean fluorescence intensity.

Close modal

In order to study if DC in coMLN differ from their counterparts in siMLN, total RNA from sorted pDCs and CD103+ DCs was sequenced and analyzed for gene expression both from coMLN and siMLN. RALDH (Aldh1a2) mRNA expression was lower in coMLN-derived CD103+ DCs compared with CD103+ DCs in siMLN, consistent with the findings of Houston et al. (18). This finding is in line with the notion that CD103+ DCs in coMLN and siMLN are not identical and that T cells are likely not imprinted with CCR9 expression in coMLN. Differences in the expression of cytokine and chemokine genes were also detected between CD103+ DC in coMLN and siMLN, and the same was true for pDC. In total, 416 (p value for individual gene <0.05) and 698 (FDR < 0.05) genes showed differential expression in CD103+ DC and pDC, respectively, when comparing either subset in coMLN and siMLN (Supplemental Table II). Comparison of coMLN-derived pDC with previously documented phenotypes of pDC revealed many markers shared between these, whereas significant differences were observed with some markers (Supplemental Table III). CoMLN-derived pDC expressed the common pDC markers PDCA-1 (BST2), B220, Siglec-H, and Ly6C, as well as pattern recognition receptor TLR7. The most striking difference was that pDCs from coMLNs and siMLN did not express IFN-α nor did they express TLR9. Expression of many other markers earlier documented in pDC were expressed also in coMLN-derived pDC, including chemokine receptors and IFN regulatory factors (IRFs). Among these, the coMLN-derived pDCs expressed IRF1 and -3 not reported to be expressed in pDCs from other tissues.

In order to confirm that pDC are activated only in coMLN during early C. rodentium infection and to study the effects of infection on both the pDC and the cDC subsets in more detail, RNA was sequenced from both DC subsets after C. rodentium infection, and gene expression was compared between infected and steady-state conditions. A total of 13,945 different transcripts were identified. Not surprisingly, pDCs and CD103+ cDCs expressed clearly different transcriptomes even under steady-state conditions (Fig. 6A, 6B, Supplemental Fig. 2). To examine the effect of C. rodentium infection on gene expression, we filtered the dataset by selecting only those transcripts that had minimum 1.5-fold change over mock infection and FDR <0.05. This resulted in 750 and 220 differentially expressed genes (DEGs) in pDCs in coMLN and siMLN, respectively (Fig. 6C, for all of the DEGs, see Supplemental Table IV). Among these were several immunologically relevant genes involved in IL-1, IL-6, and TLR signaling (Table I). Surprisingly, for cDCs, only two DEGs (Atp1b2 8.1-fold upregulation and Trav15-2-dv6-2 5.7-fold downregulation) fulfilled the same cutoff criteria in coMLN, and no genes were identified in siMLN fulfilling the same criteria for DEGs.

FIGURE 6.

C. rodentium infection modifies gene expression especially in pDCs and in colon-draining LNs. Comparison of the two DC subsets during steady-state conditions indicates a large set of genes differentially expressed between these subsets both in coMLN and siMLN (A and B). C. rodentium significantly changed the transcriptome in pDCs by activating numerous genes (C), whereas CD103+ cDCs transcriptome did not change (D) and can be demonstrated only by using less stringent criteria (see text). The effect of C. rodentium on pDCs is more pronounced in coMLN than in siMLN (C). Regression curve analysis of the effect of C. rodentium on the global transcriptome level shows that gene expression is altered widely in pDC (E) but not nearly to a similar degree in CD103+ cDCs (F). Data in (C) and (D) include all DEGs with fold change (FC) >1.5 and FDR <0.05. n = 3 with each sample pooled from four animals.

FIGURE 6.

C. rodentium infection modifies gene expression especially in pDCs and in colon-draining LNs. Comparison of the two DC subsets during steady-state conditions indicates a large set of genes differentially expressed between these subsets both in coMLN and siMLN (A and B). C. rodentium significantly changed the transcriptome in pDCs by activating numerous genes (C), whereas CD103+ cDCs transcriptome did not change (D) and can be demonstrated only by using less stringent criteria (see text). The effect of C. rodentium on pDCs is more pronounced in coMLN than in siMLN (C). Regression curve analysis of the effect of C. rodentium on the global transcriptome level shows that gene expression is altered widely in pDC (E) but not nearly to a similar degree in CD103+ cDCs (F). Data in (C) and (D) include all DEGs with fold change (FC) >1.5 and FDR <0.05. n = 3 with each sample pooled from four animals.

Close modal
Table I.
The most significant changes in gene expression in pDCs following C. rodentium infection
logFCp ValueFDRFunction/Pathway Association
Differentially expressed pDC genes in coMLN     
 Il1r2 8.31 1.1178E-06 0.00023 IL-1 signaling 
 Clec4e 8.18 0.00034 0.01160 C-type lectin involved in receptor-mediated endocytosis 
 Il1a 7.83 4.9419E-05 0.00328 Proinflammatory IL-1 family cytokine 
 Adamdec1 7.17 2.7248E-06 0.00044 Induced upon CD40-dependent maturation of DCs 
 Il6 6.72 0.00039 0.01251 IL-6 signaling 
 Il1r1 6.59 0.00025 0.00939 IL-1 signaling 
 Ikzf4 6.16 0.00149 0.02876 Ikaros family zinc finger 4; upregulated in DC stimulated by bacterial lipopeptides 
 Cd34 6.02 0.00244 0.03897 Cell adhesion (l-selectin ligand) 
 Il23r 5.78 0.00261 0.04055 DC activation 
 Cd160 5.18 9.7404E-06 0.00107 Recognizes MHC class Ia and Ib; induces proinflammatory cytokine production 
 Tlr6 4.97 8.7976E-06 0.00101 TLR signaling 
 Ngfr 4.54 0.00284 0.04283 IL-6 signaling, DC maturation 
 Irak4 2.19 1.1885E-06 0.00024 TLR signaling 
 Fas 1.91 0.00107 0.02338 Receptor-mediated cell death/activation 
 Pik3r2 1.66 2.8602E-05 0.00226 Regulatory component of PI3K 
 Map3k14 1.66 0.00254 0.03994 Binds to TRAF2 and stimulates NF-κB activity 
 Cd27 −2.07 7.2359E-05 0.00432 Supports expansion, but not effector maturation of naive T cells 
 Ccl25 −2.72 0.00260 0.04050 CCR9 ligand chemokine, attracts mucosal (α4β7) T cells 
 Mapk10 −5.36 0.00120 0.02485 MAPK 10; included in many cellular processes 
 Cxcr7 −8.20 2.0475E-07 7.1252E-05 Chemokine receptor for CXCL11 and CXCL12; MAPK activation 
 Cd163l1 −8.98 5.6214E-06 0.00072 Tissue-resident anti-inflammatory/anergic macrophages 
Differentially expressed pDC genes in siMLN     
 Il10 7.34 4.7766E-06 0.00367 IL-10, tolerogenic DC, etc. 
 Adam15 6.72 2.78E-05 0.00765 Disintegrin and metalloproteinase; Notch signaling 
 Selp 6.28 3.05E-05 0.00818 P-selectin 
 Cx3cl1 6.15 3.151E-05 0.00821 Akt signaling 
 Cd177 6.03 0.00015 0.01717 GPI-linked surface receptor on myeloid cells 
 Ccl12 5.68 0.00087 0.05025 Monocyte chemotactic protein-5, inflammatory chemokine 
 Adamdec1 5.27 3.34E-07 0.00058 Induced upon CD40-dependent maturation of DCs 
 Ccl3 5.15 3.7636E-07 0.00058 MIP-1α 
 Ccl4 3.89 9.1961E-05 0.01409 MIP-1β 
 Mapk1ip1 −2.31 0.00010 0.01423 MAPK 1–induced protein 1 
 Il2ra −2.64 8.0888E-06 0.00460 IL-2 signaling, role of JAK1 and JAK3 in γc cytokine signaling 
 Ccl17 −4.73 0.00014 0.01683 Thymus- and activation-regulated chemokine 
logFCp ValueFDRFunction/Pathway Association
Differentially expressed pDC genes in coMLN     
 Il1r2 8.31 1.1178E-06 0.00023 IL-1 signaling 
 Clec4e 8.18 0.00034 0.01160 C-type lectin involved in receptor-mediated endocytosis 
 Il1a 7.83 4.9419E-05 0.00328 Proinflammatory IL-1 family cytokine 
 Adamdec1 7.17 2.7248E-06 0.00044 Induced upon CD40-dependent maturation of DCs 
 Il6 6.72 0.00039 0.01251 IL-6 signaling 
 Il1r1 6.59 0.00025 0.00939 IL-1 signaling 
 Ikzf4 6.16 0.00149 0.02876 Ikaros family zinc finger 4; upregulated in DC stimulated by bacterial lipopeptides 
 Cd34 6.02 0.00244 0.03897 Cell adhesion (l-selectin ligand) 
 Il23r 5.78 0.00261 0.04055 DC activation 
 Cd160 5.18 9.7404E-06 0.00107 Recognizes MHC class Ia and Ib; induces proinflammatory cytokine production 
 Tlr6 4.97 8.7976E-06 0.00101 TLR signaling 
 Ngfr 4.54 0.00284 0.04283 IL-6 signaling, DC maturation 
 Irak4 2.19 1.1885E-06 0.00024 TLR signaling 
 Fas 1.91 0.00107 0.02338 Receptor-mediated cell death/activation 
 Pik3r2 1.66 2.8602E-05 0.00226 Regulatory component of PI3K 
 Map3k14 1.66 0.00254 0.03994 Binds to TRAF2 and stimulates NF-κB activity 
 Cd27 −2.07 7.2359E-05 0.00432 Supports expansion, but not effector maturation of naive T cells 
 Ccl25 −2.72 0.00260 0.04050 CCR9 ligand chemokine, attracts mucosal (α4β7) T cells 
 Mapk10 −5.36 0.00120 0.02485 MAPK 10; included in many cellular processes 
 Cxcr7 −8.20 2.0475E-07 7.1252E-05 Chemokine receptor for CXCL11 and CXCL12; MAPK activation 
 Cd163l1 −8.98 5.6214E-06 0.00072 Tissue-resident anti-inflammatory/anergic macrophages 
Differentially expressed pDC genes in siMLN     
 Il10 7.34 4.7766E-06 0.00367 IL-10, tolerogenic DC, etc. 
 Adam15 6.72 2.78E-05 0.00765 Disintegrin and metalloproteinase; Notch signaling 
 Selp 6.28 3.05E-05 0.00818 P-selectin 
 Cx3cl1 6.15 3.151E-05 0.00821 Akt signaling 
 Cd177 6.03 0.00015 0.01717 GPI-linked surface receptor on myeloid cells 
 Ccl12 5.68 0.00087 0.05025 Monocyte chemotactic protein-5, inflammatory chemokine 
 Adamdec1 5.27 3.34E-07 0.00058 Induced upon CD40-dependent maturation of DCs 
 Ccl3 5.15 3.7636E-07 0.00058 MIP-1α 
 Ccl4 3.89 9.1961E-05 0.01409 MIP-1β 
 Mapk1ip1 −2.31 0.00010 0.01423 MAPK 1–induced protein 1 
 Il2ra −2.64 8.0888E-06 0.00460 IL-2 signaling, role of JAK1 and JAK3 in γc cytokine signaling 
 Ccl17 −4.73 0.00014 0.01683 Thymus- and activation-regulated chemokine 

pDCs react differentially in coMLN and siMLN and activate different pathways. logFC (fold change) is calculated against expression level in pDC samples from mock-infected mice.

TRAF, TNFR-associated factor.

C. rodentium infection affected gene expression in pDCs clearly more profoundly in coMLN than in siMLN (Fig. 6C). Although pDCs in coMLN reacted to C. rodentium by differentially regulating 3.4 times as many genes than pDCs in siMLN (i.e., 750 versus 220 genes), only 22 upregulated genes and only 6 downregulated genes were shared among the coMLN and siMLN pDCs. In order to find any potential DEGs in CD103+ cDCs, we then employed less stringent criteria for these. Employing less stringent criteria for DEGs, CD103+ DCs up- or downregulated 147 and 54 genes in coMLN and siMLN, respectively. These included few genes directly relevant for antimicrobial immunity, namely ILs Il1b (coMLN only), Il17f, and Il22, which showed 2.7–6.9-fold upregulation after C. rodentium infection (Supplemental Fig. 3, Supplemental Table V). This suggests that C. rodentium infection may reflect some unique effects also on cDCs in coMLN, although the relatively loose criteria employed increases the risk of inadvertent differences. Comparison between pDC and CD103+ DC transcripts showed clear differences between the C-type lectin and MHC gene expression (Supplemental Table VI). Six different Clec genes were identified to be differentially expressed in the two DC populations. Clec11a, -4a, 4b, and -12a were more abundant in pDCs, whereas Clec4d and -4e were more abundant in CD103+ DCs. Furthermore, CD8α and CD8β expression showed decreasing trend (1.7- and 3-fold, respectively) after C. rodentium inoculation, indicating loss of tolerogenic pDCs in favor of immunogenic pDCs (31). However, a comparison of regression curves of gene expression between infected and mock-infected mice showed that on global gene expression level, C. rodentium infection affected gene expression in pDCs only (Fig. 6E, 6F).

In order to evaluate where the T cell response to colon-derived pathogen is initiated, we determined IFN-γ and IL-17A production in individual T cells after C. rodentium infection in coMLN and siMLN (Fig. 7). IFN-γ–producing CD4+ cells were significantly and specifically expanded in the coMLN, when compared with either mock-infected animals or the siMLN compartment (Fig. 7D). Only slight IFN-γ elevation, which was comparable to non–gut-associated brachial LN, was observed in the siMLN when compared with mock infected animals (Fig. 7D). IFN-γ–producing CD8+ cells were also expanded in response to C. rodentium infection, and this occurred significantly in both coMLN and siMLN, but not in brachial LN (Fig. 7E). In mock-infected mice, the fraction of IFN-γ–producing cells was observed to be at the same level among all LNs, remaining <2% in CD4+ cells and ranging between 5 and 13% in CD8+ cells. IL-17–producing CD4+ cells were present at considerable numbers but not significantly expanded in any LN postinfection (Fig. 7F).

FIGURE 7.

CD4 and CD8 T cells respond to C. rodentium infection in coMLNs. IFN-γ production is induced postinfection in both T cell populations. (A) Live single cells were gated. Lymphocytes were selected according to size and differentiated by CD4 and CD8 expression. (B and C) Representative dot plots are shown for IFN-γ production for CD4 and CD8 cells; numbers indicate quadrant percentages. (D) CD4 T cells show profound IFN-γ in coMLN, whereas in siMLN, the induction is very modest. (E) Both siMLNs and coMLNs show increased IFN-γ production by CD8 T cells. (F) IL-17 production is detected in a proportion of CD4 T cells already during steady-state conditions but is not induced during infection. Production of IFN-γ and IL-17 by CD4+ and CD8+ T cells was analyzed by intracellular cytokine staining on single-cell level. *p < 0.05, **p < 0.01. BLN, brachial LN; FSC-A, forward light scatter-area; SSC-A, side scatter-area.

FIGURE 7.

CD4 and CD8 T cells respond to C. rodentium infection in coMLNs. IFN-γ production is induced postinfection in both T cell populations. (A) Live single cells were gated. Lymphocytes were selected according to size and differentiated by CD4 and CD8 expression. (B and C) Representative dot plots are shown for IFN-γ production for CD4 and CD8 cells; numbers indicate quadrant percentages. (D) CD4 T cells show profound IFN-γ in coMLN, whereas in siMLN, the induction is very modest. (E) Both siMLNs and coMLNs show increased IFN-γ production by CD8 T cells. (F) IL-17 production is detected in a proportion of CD4 T cells already during steady-state conditions but is not induced during infection. Production of IFN-γ and IL-17 by CD4+ and CD8+ T cells was analyzed by intracellular cytokine staining on single-cell level. *p < 0.05, **p < 0.01. BLN, brachial LN; FSC-A, forward light scatter-area; SSC-A, side scatter-area.

Close modal

Recently, intestinal monocytes and macrophages were found to be required for Th1 polarization of Th cells in response to C. rodentium (19), but the site where naive T cells are primed and initially polarized toward Th1 polarization after C. rodentium infection has hitherto not been addressed. In this study, we describe three small LNs in the mesenterium, which drain lymph selectively from the large intestine and identify them as the site in which DC and Th1 helper T cells are activated in response to C. rodentium infection. Although cDCs showed phenotypic signs of activation earlier than pDCs, transcriptional profiling identified many more genes becoming upregulated in pDCs as compared with gut-associated (CD103+) cDCs. Genes upregulated in pDCs included genes involved in pathogen-sensing and in adaptive immune responses, suggesting an important role not only for CD103+ cDCs (19), but also for pDCs in priming protective immunity during C. rodentium infection.

It was suggested already in 1974 by Carter and Collins (16), and documented more convincingly by Van den Broeck et al. in 2006 (17), that lymphatics from different parts of the intestine drain into different parts of the cluster of MLN in the mouse. However, only very recently, Houston et al. (18) documented for the first time, to our knowledge, the existence of three LNs draining lymph specifically from the colon and showed evidence of CD103+ cDC migration from colon into these LNs. Whereas the standard method to detect lymphatic drainage to a given LN has been to inject ink such as Evan blue into the tissue of interest, we used subserosal injections of FITC-Dx into the colon wall and dissected thereafter the whole gut and mesenterium en bloc for fluorescence microscopy. This allowed the simultaneous imaging of the structure of small afferent lymphatic vessels initiating from the gut wall and identification of the collecting LN. Three separate LNs were identified draining different parts of the colon. The three separate LNs, termed in this study as coMLNs, included one at the end of siMLNs (the main MLN) closer to cecum and two at the opposite side of the central siMLNs.

As the first measure to evaluate the role of coMLNs as sentinels of infective colitis, we followed CD103+ cDC and pDC activation in local LNs after oral inoculation of C. rodentium. Together with the visual finding of enlargement of coMLNs, we verified that pDCs become activated specifically in coMLNs after C. rodentium inoculation. Interestingly, CD103+ cDC in LNs appeared to become activated earlier than pDC, but this occurred both in coMLN and siMLN with no significant changes to the transcriptome. Also, while characterizing the CD103+ cDC population, we observed the lack of CD11b+ subpopulation in the coMLN. This finding is in line with recently published results by Veenbergen et al. (32), who studied the caudal and ileac LNs (ILNs), which drain the very distal part of the colon and rectum, and reported that ILNs lack the CD11b+ migratory CD103+ DCs. Although the ILNs are separate from the coMLNs described in this study and were not analyzed in this paper, current results indicate the same phenomenon also in the coMLNs. To further evaluate the role of CD103+ cDC and pDC in coMLN, we tested their uptake of soluble Ag after subserosal injections of Alexa 488–labeled OVA into the colon wall. Unexpectedly, Alexa 488 signal was much higher in pDCs than in migratory CD103+ cDCs. pDCs have been reported to capture OVA and hen egg lysozyme both in vitro and in vivo also earlier (3335). Consistent with earlier reports (36, 37), coMLN-derived pDCs expressed lower levels of costimulatory ligands and MHC class II molecules than CD103+ cDCs. Nevertheless, pDCs are considered capable of processing and presenting Ags to initiate T cell activation (12). The levels of nonclassical MHC molecules differed between pDCs and CD103+ cDCs in coMLN. This suggests that these specialized pDCs in coMLN may have nonredundant roles in presentation of various microbial Ags to T cells, at least if they appear in the LN in a soluble form. This could occur particularly under conditions in which epithelial permeability and lymph flow are increased such as during colon inflammation. In summary, our findings of the activation kinetics of CD103+ cDCs and pDCs and the capture of soluble Ag after short-term lymph drainage are consistent with the idea, that migratory CD103+ cDCs are the first DCs to sample the bypassing and attaching C. rodentium in both small intestine and the colon. Only once the pathogen attaches firmly to colon wall and infects the mucosal layers, it gains access to coMLNs and can be taken up by resident pDCs therein.

Although rapid production of type I IFNs in response to viral nucleic acids is often regarded as the most important function of pDCs, pDCs in the small intestinal Peyer’s patches appear not to secrete IFN-α in response to TLR9 stimulation (38). Ab crosslinking studies have indicated that crosslinking pDC receptors such as BDCA-2, Siglec-H, and DC inhibitory receptor 2 strongly inhibit type I IFN production in human pDC (3941). Our RNAseq results indicated a high-level expression of Siglec-H and CLEC4A, the mouse homolog of DC inhibitory receptor 2, in coMLN-derived pDCs, suggesting that ligand binding to these could occur during C. rodentium infection and inhibit type I IFN production after pDC activation. Of note, coMLN-derived pDCs also lacked expression of TLR9, which mediates CpG-induced type I IFN production. Also, the use of transcription factors differed in coMLN pDCs, as a wider variety of IRFs were expressed in coMLN-derived pDCs in comparison with pDCs studied earlier. Together with CD103+ DC subpopulation composition in the coMLN, these LNs align more closely with the Peyer’s patches than with the siMLNs (28, 38). These data suggest specialization of coMLN-derived pDCs to detect microbial signals different from other pDCs and their adaptation to modulate gut-associated immune responses.

Along with the detailed analyses of Houston et al. (18), our findings conclusively show that immune surveillance of the small and large intestine is operated by distinct LNs and cells dedicated to these structures. The finding that pDCs upregulated expression of proinflammatory cytokine genes as well as genes involved in pathogen sensing underscores the importance of pDCs in coMLNs as active sentinels of inflammation in the colon. C-type lectins recognize a variety of microbes from fungi and viruses to bacteria and may deliver either inhibitory or activating signals to immune cells expressing them (42). In the case of C. rodentium infection, we hypothesize that lectin receptors on pDCs such as CLEC4a/e and Siglec-H may be involved either directly or indirectly in the recognition and processing of microbes by pDCs and, together with other pathogen-sensing motifs, participate in activating or regulating the immune response against C. rodentium in the coMLNs described in this study.

We thank the Cell Imaging Core for guidance with cell sorting and imaging systems.

This work was supported by the Päivikki and Sakari Sohlberg Foundation, Academy of Finland (Grants 285503 to R.T., 218057 to A.H., and Centre of Excellence in Molecular Systems Immunology and Physiology Research, 2012–2017), Finnish Diabetes Research Foundation, Maud Kuistila Memorial Fund, Turku University Foundation, Sigrid Jusélius Foundation, and Finnish Microarray and Sequencing Centre (University of Turku and Åbo Akademi University and Biocenter Finland Genome-Wide Methods Technology Platform).

The online version of this article contains supplemental material.

Abbreviations used in this article:

cDC

conventional dendritic cell

coMLN

colon-draining MLN

DC

dendritic cell

DEG

differentially expressed gene

FDR

false discovery rate

FITC-Dx

FITC-labeled dextran

ILN

ileac LN

IRF

IFN regulatory factor

LN

lymph node

MLN

mesenteric LN

pDC

plasmacytoid DC

PDCA-1

pDC Ag-1

siMLN

small intestine–draining MLN.

1
Schulz
O.
,
Jaensson
E.
,
Persson
E. K.
,
Liu
X.
,
Worbs
T.
,
Agace
W. W.
,
Pabst
O.
.
2009
.
Intestinal CD103+, but not CX3CR1+, antigen sampling cells migrate in lymph and serve classical dendritic cell functions.
J. Exp. Med.
206
:
3101
3114
.
2
Liu
L. M.
,
MacPherson
G. G.
.
1991
.
Lymph-borne (veiled) dendritic cells can acquire and present intestinally administered antigens.
Immunology
73
:
281
286
.
3
Liu
L. M.
,
MacPherson
G. G.
.
1993
.
Antigen acquisition by dendritic cells: intestinal dendritic cells acquire antigen administered orally and can prime naive T cells in vivo.
J. Exp. Med.
177
:
1299
1307
.
4
Johansson-Lindbom
B.
,
Svensson
M.
,
Pabst
O.
,
Palmqvist
C.
,
Marquez
G.
,
Förster
R.
,
Agace
W. W.
.
2005
.
Functional specialization of gut CD103+ dendritic cells in the regulation of tissue-selective T cell homing.
J. Exp. Med.
202
:
1063
1073
.
5
Coombes
J. L.
,
Siddiqui
K. R.
,
Arancibia-Cárcamo
C. V.
,
Hall
J.
,
Sun
C. M.
,
Belkaid
Y.
,
Powrie
F.
.
2007
.
A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism.
J. Exp. Med.
204
:
1757
1764
.
6
Sun
C. M.
,
Hall
J. A.
,
Blank
R. B.
,
Bouladoux
N.
,
Oukka
M.
,
Mora
J. R.
,
Belkaid
Y.
.
2007
.
Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid.
J. Exp. Med.
204
:
1775
1785
.
7
Semmrich
M.
,
Plantinga
M.
,
Svensson-Frej
M.
,
Uronen-Hansson
H.
,
Gustafsson
T.
,
Mowat
A. M.
,
Yrlid
U.
,
Lambrecht
B. N.
,
Agace
W. W.
.
2012
.
Directed antigen targeting in vivo identifies a role for CD103+ dendritic cells in both tolerogenic and immunogenic T-cell responses.
Mucosal Immunol.
5
:
150
160
.
8
Shortman
K.
,
Naik
S. H.
.
2007
.
Steady-state and inflammatory dendritic-cell development.
Nat. Rev. Immunol.
7
:
19
30
.
9
Bendriss-Vermare
N.
,
Barthélémy
C.
,
Durand
I.
,
Bruand
C.
,
Dezutter-Dambuyant
C.
,
Moulian
N.
,
Berrih-Aknin
S.
,
Caux
C.
,
Trinchieri
G.
,
Brière
F.
.
2001
.
Human thymus contains IFN-alpha-producing CD11c(-), myeloid CD11c(+), and mature interdigitating dendritic cells.
J. Clin. Invest.
107
:
835
844
.
10
Summers
K. L.
,
Hock
B. D.
,
McKenzie
J. L.
,
Hart
D. N.
.
2001
.
Phenotypic characterization of five dendritic cell subsets in human tonsils.
Am. J. Pathol.
159
:
285
295
.
11
Wendland
M.
,
Czeloth
N.
,
Mach
N.
,
Malissen
B.
,
Kremmer
E.
,
Pabst
O.
,
Förster
R.
.
2007
.
CCR9 is a homing receptor for plasmacytoid dendritic cells to the small intestine.
Proc. Natl. Acad. Sci. USA
104
:
6347
6352
.
12
Villadangos
J. A.
,
Young
L.
.
2008
.
Antigen-presentation properties of plasmacytoid dendritic cells.
Immunity
29
:
352
361
.
13
Li
H. S.
,
Gelbard
A.
,
Martinez
G. J.
,
Esashi
E.
,
Zhang
H.
,
Nguyen-Jackson
H.
,
Liu
Y. J.
,
Overwijk
W. W.
,
Watowich
S. S.
.
2011
.
Cell-intrinsic role for IFN-α-STAT1 signals in regulating murine Peyer patch plasmacytoid dendritic cells and conditioning an inflammatory response.
Blood
118
:
3879
3889
.
14
Lombardi
V. C.
,
Khaiboullina
S. F.
.
2014
.
Plasmacytoid dendritic cells of the gut: relevance to immunity and pathology.
Clin. Immunol.
153
:
165
177
.
15
Mizuno
S.
,
Kanai
T.
,
Mikami
Y.
,
Sujino
T.
,
Ono
Y.
,
Hayashi
A.
,
Handa
T.
,
Matsumoto
A.
,
Nakamoto
N.
,
Matsuoka
K.
, et al
.
2012
.
CCR9+ plasmacytoid dendritic cells in the small intestine suppress development of intestinal inflammation in mice.
Immunol. Lett.
146
:
64
69
.
16
Carter
P. B.
,
Collins
F. M.
.
1974
.
The route of enteric infection in normal mice.
J. Exp. Med.
139
:
1189
1203
.
17
Van den Broeck
W.
,
Derore
A.
,
Simoens
P.
.
2006
.
Anatomy and nomenclature of murine lymph nodes: Descriptive study and nomenclatory standardization in BALB/cAnNCrl mice.
J. Immunol. Methods
312
:
12
19
.
18
Houston
S. A.
,
Cerovic
V.
,
Thomson
C.
,
Brewer
J.
,
Mowat
A. M.
,
Milling
S.
.
2016
.
The lymph nodes draining the small intestine and colon are anatomically separate and immunologically distinct.
Mucosal Immunol.
9
:
468
478
.
19
Schreiber
H. A.
,
Loschko
J.
,
Karssemeijer
R. A.
,
Escolano
A.
,
Meredith
M. M.
,
Mucida
D.
,
Guermonprez
P.
,
Nussenzweig
M. C.
.
2013
.
Intestinal monocytes and macrophages are required for T cell polarization in response to Citrobacter rodentium.
J. Exp. Med.
210
:
2025
2039
.
20
Higgins
L. M.
,
Frankel
G.
,
Douce
G.
,
Dougan
G.
,
MacDonald
T. T.
.
1999
.
Citrobacter rodentium infection in mice elicits a mucosal Th1 cytokine response and lesions similar to those in murine inflammatory bowel disease.
Infect. Immun.
67
:
3031
3039
.
21
Bolger
A. M.
,
Lohse
M.
,
Usadel
B.
.
2014
.
Trimmomatic: a flexible trimmer for Illumina sequence data.
Bioinformatics
30
:
2114
2120
.
22
Kim
D.
,
Pertea
G.
,
Trapnell
C.
,
Pimentel
H.
,
Kelley
R.
,
Salzberg
S. L.
.
2013
.
TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions.
Genome Biol.
14
:
R36-2013-14-4-r36
.
23
Anders
S.
,
Pyl
P. T.
,
Huber
W.
.
2015
.
HTSeq--a Python framework to work with high-throughput sequencing data.
Bioinformatics
31
:
166
169
.
24
Robinson
M. D.
,
McCarthy
D. J.
,
Smyth
G. K.
.
2010
.
edgeR: a Bioconductor package for differential expression analysis of digital gene expression data.
Bioinformatics
26
:
139
140
.
25
Vong
L.
,
Pinnell
L. J.
,
Määttänen
P.
,
Yeung
C. W.
,
Lurz
E.
,
Sherman
P. M.
.
2015
.
Selective enrichment of commensal gut bacteria protects against Citrobacter rodentium-induced colitis.
Am. J. Physiol. Gastrointest. Liver Physiol.
309
:
G181
G192
.
26
Geddes
K.
,
Rubino
S. J.
,
Magalhaes
J. G.
,
Streutker
C.
,
Le Bourhis
L.
,
Cho
J. H.
,
Robertson
S. J.
,
Kim
C. J.
,
Kaul
R.
,
Philpott
D. J.
,
Girardin
S. E.
.
2011
.
Identification of an innate T helper type 17 response to intestinal bacterial pathogens.
Nat. Med.
17
:
837
844
.
27
Symonds
E. L.
,
Riedel
C. U.
,
O’Mahony
D.
,
Lapthorne
S.
,
O’Mahony
L.
,
Shanahan
F.
.
2009
.
Involvement of T helper type 17 and regulatory T cell activity in Citrobacter rodentium invasion and inflammatory damage.
Clin. Exp. Immunol.
157
:
148
154
.
28
Bogunovic
M.
,
Ginhoux
F.
,
Helft
J.
,
Shang
L.
,
Hashimoto
D.
,
Greter
M.
,
Liu
K.
,
Jakubzick
C.
,
Ingersoll
M. A.
,
Leboeuf
M.
, et al
.
2009
.
Origin of the lamina propria dendritic cell network.
Immunity
31
:
513
525
.
29
Varol
C.
,
Vallon-Eberhard
A.
,
Elinav
E.
,
Aychek
T.
,
Shapira
Y.
,
Luche
H.
,
Fehling
H. J.
,
Hardt
W. D.
,
Shakhar
G.
,
Jung
S.
.
2009
.
Intestinal lamina propria dendritic cell subsets have different origin and functions.
Immunity
31
:
502
512
.
30
Rantakari
P.
,
Auvinen
K.
,
Jäppinen
N.
,
Kapraali
M.
,
Valtonen
J.
,
Karikoski
M.
,
Gerke
H.
,
Iftakhar-E-Khuda
I.
,
Keuschnigg
J.
,
Umemoto
E.
, et al
.
2015
.
The endothelial protein PLVAP in lymphatics controls the entry of lymphocytes and antigens into lymph nodes.
Nat. Immunol.
16
:
386
396
.
31
Lombardi
V.
,
Speak
A. O.
,
Kerzerho
J.
,
Szely
N.
,
Akbari
O.
.
2012
.
CD8α⁺β⁻ and CD8α⁺β⁺ plasmacytoid dendritic cells induce Foxp3⁺ regulatory T cells and prevent the induction of airway hyper-reactivity.
Mucosal Immunol.
5
:
432
443
.
32
Veenbergen
S.
,
van Berkel
L. A.
,
du Pré
M. F.
,
He
J.
,
Karrich
J. J.
,
Costes
L. M. M.
,
Luk
F.
,
Simons-Oosterhuis
Y.
,
Raatgeep
H. C.
,
Cerovic
V.
, et al
.
2015
.
Colonic tolerance develops in the iliac lymph nodes and can be established independent of CD103+ dendritic cells.
Mucosal Immunol.
In press
.
33
de Heer
H. J.
,
Hammad
H.
,
Soullié
T.
,
Hijdra
D.
,
Vos
N.
,
Willart
M. A.
,
Hoogsteden
H. C.
,
Lambrecht
B. N.
.
2004
.
Essential role of lung plasmacytoid dendritic cells in preventing asthmatic reactions to harmless inhaled antigen.
J. Exp. Med.
200
:
89
98
.
34
Sapoznikov
A.
,
Fischer
J. A.
,
Zaft
T.
,
Krauthgamer
R.
,
Dzionek
A.
,
Jung
S.
.
2007
.
Organ-dependent in vivo priming of naive CD4+, but not CD8+, T cells by plasmacytoid dendritic cells.
J. Exp. Med.
204
:
1923
1933
.
35
Young
L. J.
,
Wilson
N. S.
,
Schnorrer
P.
,
Proietto
A.
,
ten Broeke
T.
,
Matsuki
Y.
,
Mount
A. M.
,
Belz
G. T.
,
O’Keeffe
M.
,
Ohmura-Hoshino
M.
, et al
.
2008
.
Differential MHC class II synthesis and ubiquitination confers distinct antigen-presenting properties on conventional and plasmacytoid dendritic cells.
Nat. Immunol.
9
:
1244
1252
.
36
Salio
M.
,
Palmowski
M. J.
,
Atzberger
A.
,
Hermans
I. F.
,
Cerundolo
V.
.
2004
.
CpG-matured murine plasmacytoid dendritic cells are capable of in vivo priming of functional CD8 T cell responses to endogenous but not exogenous antigens.
J. Exp. Med.
199
:
567
579
.
37
Schlecht
G.
,
Garcia
S.
,
Escriou
N.
,
Freitas
A. A.
,
Leclerc
C.
,
Dadaglio
G.
.
2004
.
Murine plasmacytoid dendritic cells induce effector/memory CD8+ T-cell responses in vivo after viral stimulation.
Blood
104
:
1808
1815
.
38
Contractor
N.
,
Louten
J.
,
Kim
L.
,
Biron
C. A.
,
Kelsall
B. L.
.
2007
.
Cutting edge: Peyer’s patch plasmacytoid dendritic cells (pDCs) produce low levels of type I interferons: possible role for IL-10, TGFbeta, and prostaglandin E2 in conditioning a unique mucosal pDC phenotype.
J. Immunol.
179
:
2690
2694
.
39
Cao
W.
,
Zhang
L.
,
Rosen
D. B.
,
Bover
L.
,
Watanabe
G.
,
Bao
M.
,
Lanier
L. L.
,
Liu
Y. J.
.
2007
.
BDCA2/Fc epsilon RI gamma complex signals through a novel BCR-like pathway in human plasmacytoid dendritic cells.
PLoS Biol.
5
:
e248
.
40
Dzionek
A.
,
Sohma
Y.
,
Nagafune
J.
,
Cella
M.
,
Colonna
M.
,
Facchetti
F.
,
Günther
G.
,
Johnston
I.
,
Lanzavecchia
A.
,
Nagasaka
T.
, et al
.
2001
.
BDCA-2, a novel plasmacytoid dendritic cell-specific type II C-type lectin, mediates antigen capture and is a potent inhibitor of interferon alpha/beta induction.
J. Exp. Med.
194
:
1823
1834
.
41
Meyer-Wentrup
F.
,
Benitez-Ribas
D.
,
Tacken
P. J.
,
Punt
C. J.
,
Figdor
C. G.
,
de Vries
I. J.
,
Adema
G. J.
.
2008
.
Targeting DCIR on human plasmacytoid dendritic cells results in antigen presentation and inhibits IFN-alpha production.
Blood
111
:
4245
4253
.
42
Sancho
D.
,
Reis e Sousa
C.
.
2012
.
Signaling by myeloid C-type lectin receptors in immunity and homeostasis.
Annu. Rev. Immunol.
30
:
491
529
.

The authors have no financial conflicts of interest.

Supplementary data