Intraepithelial lymphocytes (IELs) play an important role in maintaining the physiology of the small intestine. The majority of mouse IELs express CD8αα and are either γδ or αβ T cells. Although the development and homing of CD8αα IELs have been studied in some detail, the factors controlling their homeostasis and positioning are incompletely understood. Here we demonstrate that G protein–coupled receptor 18 (GPR18) is abundantly expressed in CD8αα IELs and that mice lacking this orphan receptor have reduced numbers of γδT IELs. Mixed bone marrow chimera experiments reveal a markedly reduced contribution of GPR18-deficient cells to the CD8αα IEL compartment and a reduction in the CD8αβ T cell subset. These defects could be rescued by transduction with a GPR18-expressing retrovirus. The GPR18-deficient γδT IELs that remained in mixed chimeras had elevated Thy1, and there were less granzyme B+ and Vγ7+ cells, indicating a greater reduction in effector-type cells. Flow cytometric analysis indicated GPR18 deficiency more strongly affected the CD8αα cells in the intraepithelial compared with the adjacent lamina propria compartment. These findings establish a requirement for GPR18 in CD8αα and CD8αβ IELs, and we suggest the receptor has a role in augmenting the accumulation of CD8 T cells in the intraepithelial versus lamina propria compartment.

Distributed along the length of the small intestine at a density of ∼1 per 10 epithelial cells, intraepithelial lymphocytes (IELs) constitute a large population of barrier immune cells (Hayday et al., 2001; Abadie et al., 2012). In mice the majority of IELs express the CD8αα homodimer, and 40–60% bear a γδTCR (Hayday et al., 2001; Cheroutre et al., 2011). γδT IELs are predominantly Vγ7+ (nomenclature of Heilig and Tonegawa [1986]), and they contribute to maintaining intestinal barrier function in the healthy state and during mucosal infections (Cheroutre et al., 2011; Abadie et al., 2012). In humans, IEL numbers increase in several conditions, including inflammatory bowel disease, and epithelial γδT lymphocytosis is a marker of celiac disease progression (Cheroutre et al., 2011; Abadie et al., 2012). γδT IELs develop from double-negative thymic precursors and undergo further maturation in the periphery before taking on a mature Thy1lo, granzyme Bhi IEL phenotype (Johansson-Lindbom and Agace, 2007; Ma et al., 2009; Guy-Grand et al., 2013). CD8αα αβT IELs develop from a unique self-reactive subset of double-positive thymocytes (Lambolez et al., 2007). The less abundant CD8αβ TCRαβ and minor CD4 TCRαβ IEL subsets represent mucosa-homing effector lymphocytes and are closely related to the main lamina propria T cell populations (Arstila et al., 2000; Cheroutre et al., 2011). CD8αα γδT and αβT IELs, but not CD8αβ IELs, are dependent on IL15 and the aryl hydrocarbon receptor (AHR), and epithelial cells are a necessary source of the trans-presenting IL15Rα chain (Abadie et al., 2012).

The intestinal epithelium is separated from the lamina propria by a basement membrane (Edelblum et al., 2012). As well as having a rich supply of blood and lymphatic vessels, the lamina propria contains T cells, dendritic cells, and plasma cells. The T cells are predominantly CD4 αβT cells, and there are very few cells with an IEL phenotype in the lamina propria (Cheroutre et al., 2011). The chemokine CCL25 (TECK) and its receptor CCR9 play a crucial role in T cell and plasma cell homing to the small intestinal intraepithelial and lamina propria compartments (Kunkel et al., 2003; Pabst et al., 2004; Stenstad et al., 2007; Wurbel et al., 2007). CCL25 is expressed by small intestinal epithelial cells, with expression being highest in the duodenum and decreasing incrementally in the jejunum and ileum (Stenstad et al., 2007). Mice lacking CCL25 or CCR9 exhibit 3- to 10-fold reductions in γδT CD8αα IELs (Wurbel et al., 2001, 2007; Uehara et al., 2002), and homing of mucosally activated effector CD8 T cells to the intestine is compromised (Stenstad et al., 2007; Wurbel et al., 2007). CCL25 and CCR9 deficiency was also shown to cause a reduction in CD8 but not CD4 T cell frequencies in the lamina propria (Wurbel et al., 2007). CD8αα αβT IEL numbers were not reduced in CCR9- or CCL25-deficient mice despite similar CCR9 expression on CD8αα γδT and αβT IELs (Wurbel et al., 2001, 2007; Uehara et al., 2002). CD8αβ and CD4 IEL numbers were also not reduced. The migration dynamics of IELs in the steady-state has recently been examined by two-photon microscopy (Chennupati et al., 2010; Edelblum et al., 2012). One study suggested the cells were mostly immobile (Chennupati et al., 2010), whereas a second provided evidence that they moved dynamically within and between intraepithelial niches (Edelblum et al., 2012). Despite these advances, the mechanisms controlling the homeostasis and positioning of the different intestinal T cell subsets remain incompletely understood.

Here we identify a role for a novel G protein–coupled receptor, GPR18, in promoting the maintenance of small intestinal CD8αα γδT, αβT, and CD8αβ IELs, and we provide evidence that this receptor favors positioning of CD8αα γδT cells in the intraepithelial versus lamina propria compartment.

GPR18 is highly expressed in lymphoid tissues and is widely expressed in immune cells, being abundant in splenic T and B cells (Fig. 1 A). Public domain gene expression data indicated two- to threefold higher expression of GPR18 in intestinal IELs compared with splenic T cells (http://www.immgen.org; http://refdic.rcai.riken.jp), and this was confirmed in quantitative PCR (QPCR) analysis of sorted cells, with expression being highest on CD8αα γδT IELs and CD8αα αβT IELs, followed by CD8αβ αβT IELs (Fig. 1 A). GPR18 transcript abundance in intestinal CD4 αβT cells, which are rare within IEL preparations (∼5%) and are much more abundant in the lamina propria (∼30%), was similar to the expression level in splenic lymphocytes (Fig. 1 A). To examine the role of GPR18 in lymphocytes, we generated GPR18-deficient mice by gene targeting. GPR18-deficient mice had normal numbers of splenic and LN αβ and γδT cells and B cells, and lymphocyte development in thymus and BM appeared normal (not depicted). Analysis of IELs showed there were similar numbers of total cells in matched adult (2–3 mo old) control and Gpr18−/− mice (Fig. 1 B). However, when the IEL subsets were examined, Gpr18−/− mice exhibited a selective deficiency in CD8αα γδT IELs (Fig. 1 C). The properties of the IEL compartment are known to change with age (Laky et al., 1997; Cheroutre et al., 2011), and analysis of a group of aged (∼7 mo old) Gpr18−/− and littermate control mice showed that there was now a significant reduction in total IEL numbers (Fig. 1 D). The CD8αα γδT IEL compartment continued to be the most affected (approximately fourfold reduced, P < 0.01), but CD8αα and CD8αβ αβT cell numbers were also reduced (approximately twofold, P < 0.05, n = 6; not depicted).

GPR18 has been suggested to promote cell chemotaxis in a Gαi protein–coupled manner to the endocannabinoid N-arachidonoyl-glycine (Kohno et al., 2006; McHugh et al., 2012). However, when WEHI231 B cells transduced with GPR18 were tested for responses to this ligand, no migration was observed, whereas the same cells transduced with several other receptors, including the endocannabinoid receptor CB2, the oxysterol receptor EBI2, the sphingosine-1-phosphate receptor S1PR1, and the chemokine receptor CXCR5, showed robust migratory responses to the appropriate ligands (Fig. 1 E and not depicted). The inability of GPR18 to respond to N-arachidonoyl-glycine was also noted in a study using β-arrestin recruitment to assess GPCR activation (Yin et al., 2009). Although we could not confirm N-arachidonoyl-glycine as a ligand for GPR18, we did observe that transduction of WEHI231 cells with GPR18 led to an approximately fivefold reduction in the CXCR4-mediated chemotactic response to CXCL12 (Fig. 1 E). This type of inhibitory effect is seen after transduction of WEHI231 cells with a range of established Gαi-coupled receptors, including CB2, EBI2, and S1PR1 (Fig. 1 E and not depicted), but does not occur when the cells are transduced with GPCRs that are established not to be Gαi coupled, including the thromboxane A2 receptor (TBXAR2) and S1PR2 (Fig. 1 E and not depicted). Based on these data, we suggest that GPR18 is effective in competing for Gαi binding and that it may therefore be able to couple to this G protein and have a pro-migratory role.

Analysis of duodenum sections for the distribution of γδT IELs showed that although reduced in frequency in the GPR18-deficient tissue, the CD8α γδT double-positive cells were distributed similarly in both types of mice, with most being in an intraepithelial location (Fig. 1 F). The migration dynamics of γδT IELs has recently been studied using real-time intravital imaging with discrepant findings regarding whether the cells are largely immobile (Chennupati et al., 2010), like dendritic epidermal T cells in the skin (Gray et al., 2011; Sumaria et al., 2011), or whether they are motile (Edelblum et al., 2012). To further examine whether IELs are motile and test whether GPR18 deficiency affected their movement dynamics, we developed a procedure to intravitally label CD8α+ IELs. Systemic injection of anti–CD8α-PE 5 h before analysis led to uniform labeling of CD8α+ cells in the epithelial and lamina propria compartments (Fig. 1 G). Using two-photon microscopy to image CD8α+ IELs from the luminal side of duodenal villi, we observed WT cells moving up and down the lateral intercellular junctions of epithelial cells, migrating between epithelial cells via the subepithelial space, and occasionally accessing the lamina propria (Fig. 1 H and Video 1). These behaviors for total CD8α+ IELs are similar to the findings of Edelblum et al. (2012) for γδT IELs using TcrdEGFP reporter mice, supporting the conclusion that IELs are motile and suggesting that the major IEL populations (CD8αα γδT and αβT) have similar movement dynamics. Analysis of GPR18-deficient mice using the same intravital CD8α-labeling procedure revealed that, at the level that could be resolved with our current imaging platform, their CD8α+ IEL migration dynamics were indistinguishable from those of the controls (Fig. 1 H and Video 2).

To further probe the stage at which GPR18 was influencing the accumulation of CD8αα γδT IELs, and to test whether the requirement was cell intrinsic, we generated mixed BM chimeras. This analysis revealed that even under conditions of competition there was no effect of GPR18 deficiency on CD4 or CD8 T cell numbers in the spleen or LNs (not depicted). However, within the IEL compartment there was a marked (∼10-fold) defect in CD8αα γδT cells and there was now also a reduction in CD8αα αβT cells (∼5-fold) and CD8αβ αβT cells (∼3-fold), whereas CD4 αβT cells were unaffected (Fig. 2 A). A comparison of the ratio of GPR18-deficient and WT γδT IELs along the length of the small intestine revealed a graded effect with the strongest defect being in the duodenum (Fig. 2 B). The basis for the greater impact of GPR18 deficiency under competitive compared with noncompetitive conditions is not known, but might indicate a reduced ability compared with WT cells to access or use a maturation signal or trophic factor that is in limited supply.

Surface phenotyping revealed that the Gpr18−/− γδT IELs in the mixed BM chimeras were enriched for Thy1hi cells and were depleted for the major γδTCR typical of intestinal IELs, Vγ7 (Fig. 2 C). The Gpr18−/− IELs were also depleted for granzyme B–high cells (Fig. 2 C). The high Thy1 and low granzyme B expression is consistent with a less mature effector state (Ma et al., 2009; Cheroutre et al., 2011). CD103 (αE integrin) expression was comparable on the Gpr18−/− and control cells, whereas CCR9 and CXCR3 surface levels were slightly elevated (Fig. 2 C and not depicted). Contrary to this elevated CCR9 and CXCR3 expression, GPR18-deficient γδT IELs in mixed BM chimeras showed reduced migration to the CCR9 ligand, CCL25, and the CXCR3 ligand, CXCL10 (Fig. 2 D). However, they also had a significantly reduced baseline migration in the absence of chemokine (Fig. 2 D). When the fold change in migration compared with “nil” was determined, there was an increase in the relative CCL25 response of the Gpr18−/− cells (Fig. 2 D). The alterations in CD8αα γδT cell surface phenotype and migration behavior observed for Gpr18−/− cells from mixed BM chimeras (Fig. 2, C and D) were not seen for CD8αα γδT cells from Gpr18−/− mice (Fig. 2 D and not depicted). This may indicate that these phenotypes are secondary consequences of a role played by GPR18 in helping cells to compete for a maturation signal.

CCR9-deficient mice suffer a deficiency in CD8αα γδT IEL numbers (Wurbel et al., 2001, 2007; Uehara et al., 2002) that is more severe than observed in GPR18-deficient mice (Fig. 2 E and Fig. 1 C). Analysis of CCR9 GPR18 double-deficient mice showed a similar magnitude of CD8αα γδT IEL deficiency to that observed in Ccr9−/− mice (Fig. 2 E). These data suggest the function of CCR9 is dominant over the role of GPR18 in establishing an IEL compartment of normal size. However, in contrast to Gpr18−/− γδT IELs in mixed BM chimeras (Fig. 2 C), Ccr9−/− γδT IELs in mixed BM chimeras contained normal Vγ7+ cell frequencies (Fig. 2 F). These findings indicate that GPR18 has a function distinct from or in addition to any role it might have in supporting CCR9 function.

Analysis of cytokine production after in vitro activation of cells from mixed BM chimeras revealed similar IFN-γ expression by Gpr18−/− and internal control cells but elevated TNF production by the Gpr18−/− cells (Fig. 2 G). The TNF expression level was similar to that observed in Thy1hi WT cells (not depicted), suggesting that it was secondary to the effect of GPR18 deficiency on cell maturation state in competitive chimeras. To examine the IEL turnover rate, mixed BM chimeras were maintained on drinking water containing BrdU for 1 wk and then examined by flow cytometry. Similar fractions of control and Gpr18−/− cells were BrdU labeled (Fig. 2 H), suggesting that the reduced Gpr18−/− cell number was not a consequence of a markedly accelerated loss of IELs. Consistent with this conclusion, the fraction of apoptotic γδT IELs detected by annexin V staining was similar for the mutant and WT cells (Fig. 2 I).

To confirm that the deficiency in IELs was solely caused by a loss of GPR18 and not caused by altered expression of a linked gene or other off-target effects, GPR18 expression was rescued in Gpr18−/− BM by retroviral transduction. Analysis of spleen cells from mice reconstituted with GPR18-encoding versus control retrovirus–transduced BM showed that reporter-positive cells were present at similar frequencies in both groups, confirming that GPR18 does not influence the development of recirculating T cell populations (not depicted). However, within the intestine, each of the IEL subsets that were deficient in the absence of GPR18 was markedly rescued by GPR18 transduction (Fig. 3 A). This was evident both as a higher frequency of transduced (reporter positive) cells and an increase in their number to within twofold of the normal range (Fig. 3 B and Fig. 1, B and C). Moreover, the rescued CD8αα γδT cells down-regulated Thy1 to a similar extent as control cells (Fig. 3 C).

Vγ7+ IELs are generated in the thymus and may undergo a final maturation process in mesenteric LNs (mLNs; Guy-Grand et al., 2013). Production of Vγ7+ γδT cells in the thymus of mixed BM chimeras was not affected by GPR18 deficiency (Fig. 4 A and not depicted). The cells were also present in frequencies comparable with those of Gpr18+/− controls in the mLNs (Fig. 4 A and not depicted). To test whether homing of IELs from blood to intestine might require GPR18, a mixture of total IELs from GPR18-deficient (CD45.1/2) and WT (CD45.1) donors were transferred to γδT IEL-deficient (Ccr9−/− CD45.2) recipients. Although CD8αα IELs are nonrecirculatory and home only inefficiently back to the small intestine after i.v. transfer (Hayday et al., 2001; Chennupati et al., 2010; Cheroutre et al., 2011), analysis at 1 d showed that Gpr18−/− cells were present among the transferred cells within the IEL compartment at a similar frequency to their representation in blood (Fig. 4 B). These data disfavor a role for GPR18 in promoting CD8αα IEL recruitment from blood into tissue. Given that GPR18 is thought to most likely respond to a small molecule ligand (Inoue et al., 2012), these findings are not unexpected as the step of lymphocyte recruitment from blood into tissue usually involves a protein (chemokine) ligand that can be displayed on the endothelium (Islam and Luster, 2012). CCR9 and CCL25 are established to play an important role in this recruitment step for CD8 T cells and plasma cells (Kunkel et al., 2003; Pabst et al., 2004; Stenstad et al., 2007; Wurbel et al., 2007), a requirement we could confirm for CD8 T cells that had been activated under CCR9-inducing conditions (Fig. 4 C).

CD8αα IELs are considered to be restricted to the intraepithelial compartment, and consistent with this view, the cells make up only a very minor component of standard lamina propria lymphocyte (LPL) preparations (Fig. 5 A; Cheroutre et al., 2011), and it has so far not been possible to determine their distribution in the lamina propria by immunofluorescence analysis. However, because our intravital imaging and that of Edelblum et al. (2012) suggested that CD8α+ IELs occasionally access the lamina propria (Videos 1 and 2), we asked whether GPR18 differentially affected the frequency of CD8αα γδT cells in these compartments. Analysis of IEL and LPL preparations from mixed BM chimeras revealed that the fraction of CD8αα γδT and αβT IELs was more strongly reduced in the IEL compartment than within the LPL compartment (Fig. 5 B). Reciprocally, in mice reconstituted with GPR18-tranduced Gpr18−/− BM, the reporter-positive CD8αα γδT cells were enriched in the IEL compared with the LPL compartment (Fig. 5 C). These observations suggest that GPR18 augments access of CD8αα γδT cells to, or retention within, the IEL compartment.

In summary, we establish that the orphan receptor, GPR18, is required for the normal homeostasis of CD8αα γδT and αβT IELs and CD8αβ IELs. Our intravital imaging helps resolve a discrepancy between two recent studies (Chennupati et al., 2010; Edelblum et al., 2012) by providing additional evidence that IELs are motile and that they do not remain fixed between one set of epithelial cells but travel between intraepithelial niches through a path that sometimes takes them into the lamina propria. Studies on other cell types, such as germinal center and marginal zone B cells (Allen et al., 2004; Arnon et al., 2011), indicate that for cells to move repeatedly between adjacent compartments, they need to respond to at least two GPCR ligands that are distributed unequally between the compartments. The only directional cue previously established to promote IEL positioning in the small intestine is CCL25 and its receptor CCR9, and at steady-state these studies only found effects on γδT IELs (Abadie et al., 2012; Islam and Luster, 2012). Based on our findings here, we suggest that GPR18 may respond to a ligand that is distributed in areas overlapping with but not identical to the distribution of CCL25 and that the receptor helps achieve fine control of IEL distribution within the small intestine. We suggest that by affecting the efficiency of access to currently imprecisely defined intestinal niches, GPR18 influences exposure of IELs to factors necessary for their maturation or trophic support and that the reduced ability to access these factors most strongly affects their numbers under conditions of competition. Strong competitive effects on cell numbers have been observed in other instances where trophic support is limited (e.g., Lesley et al., 2004; Thien et al., 2004). The altered maturation state may in turn be responsible for the reduced in vitro motility of the cells, an alteration which might amplify effects of GPR18 deficiency on cell positioning and homeostasis. It is notable that mice lacking IL15Rα or AHR have decreased CD8αα IELs but retain normal numbers of CD8αβ IELs (Lodolce et al., 1998; Ma et al., 2009; Li et al., 2011). The reduced competitiveness of GPR18-deficient CD8αβ as well as CD8αα IELs indicates GPR18 may be required for accessing factors in addition to or other than IL15 or AHR ligands or that it is instead required for retention of IELs in the intestine. We do not exclude the possibility that GPR18 transmits signals that have their own trophic or maturational influence on IELs. It will be important in future studies to define the nature of the GPR18 ligand and to study whether GPR18 and GPR18 ligand abundance are altered in disease states where γδT IEL numbers and distribution are affected, such as in inflammatory bowel disease and celiac disease (Cheroutre et al., 2011; Abadie et al., 2012).

Mice.

C57BL/6J (B6, CD45.2) and congenic B6 CD45.1+ mice were obtained from the Jackson Laboratory, and these strains were intercrossed to generate B6 CD45.1/2 F1 mice. CCR9-deficient mice (Uehara et al., 2002) were obtained from P. Love (National Institutes of Health, Bethesda, MD) and provided by E. Verdin (University of California, San Francisco [UCSF], San Francisco, CA). GPR18-deficient mice were generated by Ozgene Pty Ltd. using the Cre/loxP system. In brief, three DNA fragments (a 4-kb 5′ homology arm, a 1.4-kb floxed arm containing the single Gpr18 exon, and a 4-kb 3′ homology arm) were generated by nested PCR from C57BL/6 mouse genomic DNA and cloned into the pOzIII vector (Ozgene Pty Ltd.). The targeting vector was linearized and electroporated into B6-derived embryonic stem cells. Positive clones were screened and confirmed with Southern blot. Chimeras were bred to B6 mice, and germline transmission was confirmed by allele-specific PCR. Gpr18 floxed mice were intercrossed to actin-Cre transgenic mice to obtain Gpr18 germline deleted mice. To generate BM chimeras, CD45.1+ B6 mice were irradiated by exposure to 1,100 rad of γ-irradiation in two doses 3 h apart and i.v. injected with 2 × 106 total BM cells from CD45.1/2 WT plus 2 × 106 total BM cells from CD45.2 mice of each genotype as indicated (Gpr18+/−, Gpr18−/−, or Ccr9−/−) and analyzed after 2–3 mo. All chimeras appeared healthy at the time of analysis. Although the input cells were mixed at a 1:1 ratio, the reconstitution efficiencies of the different donor BMs were not always identical, a common occurrence in mixed BM chimera studies. Animals were housed in a specific pathogen–free environment in the Laboratory Animal Research Center at the UCSF, and all experiments conformed to ethical principles and guidelines approved by the UCSF Institutional Animal Care and Use Committee.

Cell preparations, adoptive transfer, and migration assays.

Thymocyte, splenocyte, and LN cell suspensions were prepared by mashing the organs through 70-µm cell strainers. IELs and LPLs were isolated as described previously (Jiang et al., 2013), with modifications. In brief, Peyer’s patches were removed, and then the small intestine was opened longitudinally and washed with PBS containing 0.1% BSA, 100 U/ml penicillin, and 100 µg/ml streptomycin three times. The intestines were then shaken with prewarmed DMEM containing penicillin, streptomycin, and 5% FCS for 30 min at 225 rpm, 37°C. Supernatants were separated on a 30–40–80% Percoll density gradient (GE Healthcare), and the cells that layered between the 40–80% fractions were collected as IELs. After IEL isolation, tissues were shaken in RPMI 1640 containing 5 mM EDTA and 5% FCS at 175 rpm, 37°C for 15 min. This step was repeated four times and the supernatants were discarded. LPLs were then isolated after digestion in RPMI-1640 supplemented with 0.5 mg/ml collagenase type II (Worthington Biochemical Corporation), 0.1 mg/ml DNase I (Sigma-Aldrich), and 10% FCS at 200 rpm, 37°C for 30 min. Released cells were then subjected to Percoll fractionation as described above for isolation of IELs. For IEL adoptive transfer, freshly isolated 3–5 million IELs of each genotype were i.v. co-transferred into recipients. For transfers of T cells activated to induce CCR9 expression, T cells from LNs of each genotype were stimulated with plate-bound CD3 and CD28 plus 100 ng/ml retinoic acid for 3 d and then resuspended in complete RPMI-1640 media with 100 U/ml IL-2 plus 100 ng/ml retinoic acid. At day 5, cells were harvested and transferred into WT recipients. Transwell migration assays were with 5-µm transwells and were performed as described previously (Ngo et al., 1998) using 106 IELs prepared by the Percoll density gradient method above. CCL25 and CXCL10 were obtained from BioLegend; CXCL12 was obtained from PeproTech; N-arachidonoyl-glycine and 2-arachidonyl-glycerol were obtained from Enzo Life Science and Sigma-Aldrich, respectively.

Antibodies and flow cytometry.

Cells were stained using standard procedures for surface markers. The following monoclonal antibodies were used for flow cytometry: TCRγδ (GL3; BD or BioLegend), TCRβ (H57; BioLegend), CD4 (GK1.5; BioLegend), CD8α (53.6.7; Tonbo Bio), CD8β (H35; eBioscience), Thy1.2 (30-H12; BD), Granzyme B (GB11; Invitrogen), CD45.1 (A20; BioLegend), and CD45.2 (104; BioLegend). Vγ7 antibody was provided by P. Pereira (Institut Pasteur, Paris, France). CD8αα cells were gated as CD8α+CD8β. For cytokine analysis, freshly isolated IELs were stimulated with 40 ng/ml PMA (Sigma-Aldrich) and 4 µg/ml ionomycin (Sigma-Aldrich) for 4.5 h in the presence of GolgiPlug (BD) in complete RPMI-1640 media. Cells were then intracellularly stained for IFN-γ (XMG1.2; BioLegend) and TNF (MP6-XT22; eBioscience) with Cytoperm/Cytofix reagents (BD) according to the manufacturer’s instructions. For BrdU incorporation analysis, mice were given water containing 0.5 mg/ml BrdU for 7 d. Staining was performed with a BrdU flow kit (BD) according to the manufacturer’s instructions. Annexin V staining of fresh IELs was performed with the Annexin V staining kit (BD).

Immunofluorescence.

Cryosections of 7 µm were fixed and stained as previously described (Wang et al., 2011) with the following first antibodies: Laminin (Invitrogen), CD8α (53.6.7; Tonbo Bio), TCRγδ (GL3; BD or BioLegend), and E-cadherin (GG3A; BD). Images were all acquired with AxioVision using an Axio Observer Z1 microscope (Carl Zeiss).

Intravital two-photon microscopy.

Mice were i.v. injected with 10 µg anti–CD8α-PE (BioLegend) 5 h before imaging. After anesthetization, mice were injected i.v. with Hoechst 33342 dye (Invitrogen), and the distal duodenum was exposed and opened along the antimesenteric border, as described previously (Edelblum et al., 2012). The mucosal surface was placed against a coverslip bottom of a 3D-printed cassette containing PBS. Images were acquired with ZEN2012 (Carl Zeiss) using a 7MP two-photon microscope (Carl Zeiss) equipped with a Chameleon laser (Coherent). Excitation wavelength was 880 nm. Images were acquired by taking 21-µm Z-stacks at 3-µm steps every 20 s. Each XY plane spans 512 × 512 µm2. Videos were made and analyzed with Imaris 7.4 ×64 (Bitplane).

Online supplemental material.

Videos 1 and 2 show the real-time imaging of CD8α+ IEL migration in Gpr18+/− and Gpr18−/− duodenum, respectively.

Thanks to Jinping An for expert help with the mouse colony, Ying Xu for QPCR analysis, Jagan Muppidi for help with epitope tagging, Hsin Chen for imaging help, and Steven Rosen, Tangsheng Yi, Hayakazu Sumida and Michael Barnes for helpful discussions.

X. Wang is an Associate and J.G. Cyster is an Investigator of the Howard Hughes Medical Institute. This work was supported in part by National Institutes of Health grant AI40098.

The authors declare no competing financial interests.

Abadie
,
V.
,
V.
Discepolo
, and
B.
Jabri
.
2012
.
Intraepithelial lymphocytes in celiac disease immunopathology
.
Semin. Immunopathol.
34
:
551
566
.
Allen
,
C.D.
,
K.M.
Ansel
,
C.
Low
,
R.
Lesley
,
H.
Tamamura
,
N.
Fujii
, and
J.G.
Cyster
.
2004
.
Germinal center dark and light zone organization is mediated by CXCR4 and CXCR5
.
Nat. Immunol.
5
:
943
952
.
Arnon
,
T.I.
,
Y.
Xu
,
C.
Lo
,
T.
Pham
,
J.
An
,
S.
Coughlin
,
G.W.
Dorn
, and
J.G.
Cyster
.
2011
.
GRK2-dependent S1PR1 desensitization is required for lymphocytes to overcome their attraction to blood
.
Science.
333
:
1898
1903
.
Arstila
,
T.
,
T.P.
Arstila
,
S.
Calbo
,
F.
Selz
,
M.
Malassis-Seris
,
P.
Vassalli
,
P.
Kourilsky
, and
D.
Guy-Grand
.
2000
.
Identical T cell clones are located within the mouse gut epithelium and lamina propia and circulate in the thoracic duct lymph
.
J. Exp. Med.
191
:
823
834
.
Chennupati
,
V.
,
T.
Worbs
,
X.
Liu
,
F.H.
Malinarich
,
S.
Schmitz
,
J.D.
Haas
,
B.
Malissen
,
R.
Förster
, and
I.
Prinz
.
2010
.
Intra- and intercompartmental movement of γδ T cells: intestinal intraepithelial and peripheral γδ T cells represent exclusive nonoverlapping populations with distinct migration characteristics
.
J. Immunol.
185
:
5160
5168
.
Cheroutre
,
H.
,
F.
Lambolez
, and
D.
Mucida
.
2011
.
The light and dark sides of intestinal intraepithelial lymphocytes
.
Nat. Rev. Immunol.
11
:
445
456
.
Edelblum
,
K.L.
,
L.
Shen
,
C.R.
Weber
,
A.M.
Marchiando
,
B.S.
Clay
,
Y.
Wang
,
I.
Prinz
,
B.
Malissen
,
A.I.
Sperling
, and
J.R.
Turner
.
2012
.
Dynamic migration of γδ intraepithelial lymphocytes requires occludin
.
Proc. Natl. Acad. Sci. USA.
109
:
7097
7102
.
Gray
,
E.E.
,
K.
Suzuki
, and
J.G.
Cyster
.
2011
.
Cutting edge: Identification of a motile IL-17-producing γδ T cell population in the dermis
.
J. Immunol.
186
:
6091
6095
.
Guy-Grand
,
D.
,
P.
Vassalli
,
G.
Eberl
,
P.
Pereira
,
O.
Burlen-Defranoux
,
F.
Lemaitre
,
J.P.
Di Santo
,
A.A.
Freitas
,
A.
Cumano
, and
A.
Bandeira
.
2013
.
Origin, trafficking, and intraepithelial fate of gut-tropic T cells
.
J. Exp. Med.
210
:
1839
1854
.
Hayday
,
A.
,
E.
Theodoridis
,
E.
Ramsburg
, and
J.
Shires
.
2001
.
Intraepithelial lymphocytes: exploring the Third Way in immunology
.
Nat. Immunol.
2
:
997
1003
.
Heilig
,
J.S.
, and
S.
Tonegawa
.
1986
.
Diversity of murine gamma genes and expression in fetal and adult T lymphocytes
.
Nature.
322
:
836
840
.
Inoue
,
A.
,
J.
Ishiguro
,
H.
Kitamura
,
N.
Arima
,
M.
Okutani
,
A.
Shuto
,
S.
Higashiyama
,
T.
Ohwada
,
H.
Arai
,
K.
Makide
, and
J.
Aoki
.
2012
.
TGFα shedding assay: an accurate and versatile method for detecting GPCR activation
.
Nat. Methods.
9
:
1021
1029
.
Islam
,
S.A.
, and
A.D.
Luster
.
2012
.
T cell homing to epithelial barriers in allergic disease
.
Nat. Med.
18
:
705
715
.
Jiang
,
W.
,
X.
Wang
,
B.
Zeng
,
L.
Liu
,
A.
Tardivel
,
H.
Wei
,
J.
Han
,
H.R.
MacDonald
,
J.
Tschopp
,
Z.
Tian
, and
R.
Zhou
.
2013
.
Recognition of gut microbiota by NOD2 is essential for the homeostasis of intestinal intraepithelial lymphocytes
.
J. Exp. Med.
210
:
2465
2476
.
Johansson-Lindbom
,
B.
, and
W.W.
Agace
.
2007
.
Generation of gut-homing T cells and their localization to the small intestinal mucosa
.
Immunol. Rev.
215
:
226
242
.
Kohno
,
M.
,
H.
Hasegawa
,
A.
Inoue
,
M.
Muraoka
,
T.
Miyazaki
,
K.
Oka
, and
M.
Yasukawa
.
2006
.
Identification of N-arachidonylglycine as the endogenous ligand for orphan G-protein-coupled receptor GPR18
.
Biochem. Biophys. Res. Commun.
347
:
827
832
.
Kunkel
,
E.J.
,
D.J.
Campbell
, and
E.C.
Butcher
.
2003
.
Chemokines in lymphocyte trafficking and intestinal immunity
.
Microcirculation.
10
:
313
323
.
Laky
,
K.
,
L.
Lefrançois
, and
L.
Puddington
.
1997
.
Age-dependent intestinal lymphoproliferative disorder due to stem cell factor receptor deficiency: parameters in small and large intestine
.
J. Immunol.
158
:
1417
1427
.
Lambolez
,
F.
,
M.
Kronenberg
, and
H.
Cheroutre
.
2007
.
Thymic differentiation of TCRαβ+ CD8αα+ IELs
.
Immunol. Rev.
215
:
178
188
.
Lesley
,
R.
,
Y.
Xu
,
S.L.
Kalled
,
D.M.
Hess
,
S.R.
Schwab
,
H.B.
Shu
, and
J.G.
Cyster
.
2004
.
Reduced competitiveness of autoantigen-engaged B cells due to increased dependence on BAFF
.
Immunity.
20
:
441
453
.
Li
,
Y.
,
S.
Innocentin
,
D.R.
Withers
,
N.A.
Roberts
,
A.R.
Gallagher
,
E.F.
Grigorieva
,
C.
Wilhelm
, and
M.
Veldhoen
.
2011
.
Exogenous stimuli maintain intraepithelial lymphocytes via aryl hydrocarbon receptor activation
.
Cell.
147
:
629
640
.
Lodolce
,
J.P.
,
D.L.
Boone
,
S.
Chai
,
R.E.
Swain
,
T.
Dassopoulos
,
S.
Trettin
, and
A.
Ma
.
1998
.
IL-15 receptor maintains lymphoid homeostasis by supporting lymphocyte homing and proliferation
.
Immunity.
9
:
669
676
.
Ma
,
L.J.
,
L.F.
Acero
,
T.
Zal
, and
K.S.
Schluns
.
2009
.
Trans-presentation of IL-15 by intestinal epithelial cells drives development of CD8αα IELs
.
J. Immunol.
183
:
1044
1054
.
McHugh
,
D.
,
J.
Wager-Miller
,
J.
Page
, and
H.B.
Bradshaw
.
2012
.
siRNA knockdown of GPR18 receptors in BV-2 microglia attenuates N-arachidonoyl glycine-induced cell migration
.
J. Mol. Signal.
7
:
10
.
Ngo
,
V.N.
,
H.L.
Tang
, and
J.G.
Cyster
.
1998
.
Epstein-Barr virus-induced molecule 1 ligand chemokine is expressed by dendritic cells in lymphoid tissues and strongly attracts naive T cells and activated B cells
.
J. Exp. Med.
188
:
181
191
.
Pabst
,
O.
,
L.
Ohl
,
M.
Wendland
,
M.A.
Wurbel
,
E.
Kremmer
,
B.
Malissen
, and
R.
Förster
.
2004
.
Chemokine receptor CCR9 contributes to the localization of plasma cells to the small intestine
.
J. Exp. Med.
199
:
411
416
.
Stenstad
,
H.
,
M.
Svensson
,
H.
Cucak
,
K.
Kotarsky
, and
W.W.
Agace
.
2007
.
Differential homing mechanisms regulate regionalized effector CD8αβ+ T cell accumulation within the small intestine
.
Proc. Natl. Acad. Sci. USA.
104
:
10122
10127
.
Sumaria
,
N.
,
B.
Roediger
,
L.G.
Ng
,
J.
Qin
,
R.
Pinto
,
L.L.
Cavanagh
,
E.
Shklovskaya
,
B.
Fazekas de St Groth
,
J.A.
Triccas
, and
W.
Weninger
.
2011
.
Cutaneous immunosurveillance by self-renewing dermal γδ T cells
.
J. Exp. Med.
208
:
505
518
.
Thien
,
M.
,
T.G.
Phan
,
S.
Gardam
,
M.
Amesbury
,
A.
Basten
,
F.
Mackay
, and
R.
Brink
.
2004
.
Excess BAFF rescues self-reactive B cells from peripheral deletion and allows them to enter forbidden follicular and marginal zone niches
.
Immunity.
20
:
785
798
.
Uehara
,
S.
,
A.
Grinberg
,
J.M.
Farber
, and
P.E.
Love
.
2002
.
A role for CCR9 in T lymphocyte development and migration
.
J. Immunol.
168
:
2811
2819
.
Wang
,
X.
,
B.
Cho
,
K.
Suzuki
,
Y.
Xu
,
J.A.
Green
,
J.
An
, and
J.G.
Cyster
.
2011
.
Follicular dendritic cells help establish follicle identity and promote B cell retention in germinal centers
.
J. Exp. Med.
208
:
2497
2510
.
Wurbel
,
M.A.
,
M.
Malissen
,
D.
Guy-Grand
,
E.
Meffre
,
M.C.
Nussenzweig
,
M.
Richelme
,
A.
Carrier
, and
B.
Malissen
.
2001
.
Mice lacking the CCR9 CC-chemokine receptor show a mild impairment of early T- and B-cell development and a reduction in T-cell receptor γδ+ gut intraepithelial lymphocytes
.
Blood.
98
:
2626
2632
.
Wurbel
,
M.A.
,
M.
Malissen
,
D.
Guy-Grand
,
B.
Malissen
, and
J.J.
Campbell
.
2007
.
Impaired accumulation of antigen-specific CD8 lymphocytes in chemokine CCL25-deficient intestinal epithelium and lamina propria
.
J. Immunol.
178
:
7598
7606
.
Yin
,
H.
,
A.
Chu
,
W.
Li
,
B.
Wang
,
F.
Shelton
,
F.
Otero
,
D.G.
Nguyen
,
J.S.
Caldwell
, and
Y.A.
Chen
.
2009
.
Lipid G protein-coupled receptor ligand identification using β-arrestin PathHunter assay
.
J. Biol. Chem.
284
:
12328
12338
.

Abbreviations used:
AHR

aryl hydrocarbon receptor

IEL

intraepithelial lymphocyte

LPL

lamina propria lymphocyte

mLN

mesenteric LN

QPCR

quantitative PCR

Author notes

*

X. Wang’s present address is Dept. of Immunology, Nanjing Medical University, Nanjing 210029, China.

This article is distributed under the terms of an Attribution–Noncommercial–Share Alike–No Mirror Sites license for the first six months after the publication date (see http://www.rupress.org/terms). After six months it is available under a Creative Commons License (Attribution–Noncommercial–Share Alike 3.0 Unported license, as described at http://creativecommons.org/licenses/by-nc-sa/3.0/).

Supplementary data