SUMMARY
The Aryl hydrocarbon receptor (Ahr) regulates the differentiation and function of CD4+ T cells; however, its cell-intrinsic role in CD8+ T cells remains elusive. Herein we show that Ahr acts as a promoter of resident memory CD8+ T cell (TRM) differentiation and function. Genetic ablation of Ahr in mouse CD8+ T cells leads to increased CD127−KLRG1+ short-lived effector cells and CD44+CD62L+ T central memory cells but reduced granzyme-B-producing CD69+CD103+ TRM cells. Genome-wide analyses reveal that Ahr suppresses the circulating while promoting the resident memory core gene program. A tumor resident polyfunctional CD8+ T cell population, revealed by single-cell RNA-seq, is diminished upon Ahr deletion, compromising anti-tumor immunity. Human intestinal intraepithelial CD8+ T cells also highly express AHR that regulates in vitro TRM differentiation and granzyme B production. Collectively, these data suggest that Ahr is an important cell-intrinsic factor for CD8+ T cell immunity.
In brief
Dean et al. show that Ahr, an environmental sensor, is important for the development and function of a specific immune cell type called tissue resident memory CD8+ T cells. These cells are critical in the response to local infections and tumors.
Graphical Abstract
INTRODUCTION
The continuous exposure to various commensals creates a unique “physiological inflammation state” in the gut. Recently, it has been shown that treating “steady-state” specific pathogen-free mice with an antiviral cocktail led to an intestinal intraepithelial (IEL)-specific reduction of most T cell subsets including CD8αβ T cells (hereinafter referred to as CD8+ T cells).1 These IEL CD8+ T cells are CD69+CD103+ and known as T resident memory (TRM) cells.2 The lamina propria is home to a diverse population of other CD8+ T cell subsets including CD69+CD103+ CD8+ T cells.3 Following the recognition of a foreign commensal or pathogen, professional antigen-presenting cells migrate to draining lymph nodes and activate naive CD8+ T cells. Activation of naive CD8+ T cells results in clonal expansion and differentiation into diverse subsets of effector and memory cytotoxic T cell lymphocytes (CTLs).4,5 This coordinated response helps to eliminate the pathogen and provide long-term protection.
CTLs vary in their phenotype, function, proliferative capacity, longevity, and ability to differentiate into cells conferring long-lived memory.6 Cells generated during the early phase of expansion likely have a spectrum of phenotypes arising via asymmetric cell division and/or effector cell de-differentiation.7–10 Killer cell lectin-like receptor subfamily G, member 1 (KLRG1) and IL-7Rα (CD127), have been used as markers to distinguish effector CD8+ T cell subsets based on differences in migration, long-term survival, and memory differentiation potential. KLRG1+ cells, double-positive effector cells (CD127+KLRG1+, TDPE), and short-lived effector cells (CD127−KLRG1+, TSLE) cells, while being valuable functional effectors that produce cytolytic molecules such as perforin and granzyme B, at levels similar to other effector subsets,11 have been described to exhibit limited memory cell differentiation potential.12–14 In comparison, KLRG1− cells, double-negative early effector cells (KLRG1−CD127−, TEE) and memory precursor effector cells (KLRG1−CD127+, TMPE) cells demonstrate better survival during the contraction phase and maintain stemness, as they are able to differentiate into multiple memory cell lineages.13–15 This delineation is the canonical model for CD8+ T cell terminal effector vs. memory precursor differentiation; however, recent evidence suggests that there is more nuance and heterogeneity evident at this stage.16–18
Memory T cell subsets that survive the contraction phase of infection vary based on their localization and function.19 T central memory (TCM) cells express the lymph node homing receptors CCR7 and CD62L and have a high proliferative capacity but exhibit low cytotoxicity, while T effector memory (TEM) cells are highly cytotoxic but are less proliferative, do not express CCR7 and CD62L, and therefore survey non-lymphoid tissues in the case of pathogen re-exposure.20–22 In recent years, TRM cells have gained significant attention because of their ability to remain in the tissue constantly surveilling in case of re-exposure to the same pathogen.23–25 While the exact ontogeny of TRM cells is not precisely known, the precursor cells poised to differentiate into TRM have been suggested to resemble the classical TMPE phenotype, and CD103 may function as a marker of this precursor population.3,13,26 However, it has also been shown using a Klrg1-Cre fate mapping reporter mouse model that a number of cells that previously expressed KLRG1 and subsequently downregulate it (“ex-KLRG1” cells) give rise to all memory T cell lineages including TRM cells.16 Runx3, Blimp1, and Hobbit are known transcriptional regulators of the TRM lineage.27 Runx3 is essential for the differentiation and long-term maintenance of CD8+ TRM cells, while Blimp1 and Hobbit function to suppress Klf2, a transcriptional regulator important for T cell trafficking by promoting expression of S1P1 and CD62L and the TCM cell lineage.28–30 Upon secondary recognition of cognate antigen, memory cells are able to respond faster than a primary response, and it has recently been show that TRM cells, after re-infection, can leave the tissue and contribute to the systemic immune responses by differentiating into a diverse pool of effector and memory cells.31,32
The aryl hydrocarbon receptor (Ahr), a ligand-dependent environmental sensor and transcription factor, can be activated by xenobiotic compounds as well as natural ligands from diet and/or the microbiota.33,34 Before its role in regulation of the immune system was appreciated, Ahr was shown to mediate the conversion and carcinogenic effects of environmental toxins like 2,3,7,8-tetrachloro-dibenzo-p-dioxin (TCDD). More recently, a physiological role for Ahr has emerged in the regulation of the immune system development and function via ligands from the microbiome, diet, and host cell metabolism. Compared with well-studied functions in CD4+ and CD8αα T cells,33,35 the cell-intrinsic role and direct targets of Ahr in CD8+ T cell immune responses remain elusive.
RESULTS
Ahr is expressed by intestinal resident CD8αβ T cells
To more readily detect Ahr expression in different cell populations, we utilized AhrdCAIR mice,36 in which a GFP reporter was knocked into the endogenous Ahr locus under the control of Ahr cis-acting regulatory elements. Comparing different tissues, we found that small intestine (SI) intraepithelial lymphocyte (IEL), as well as lamina propria lymphocyte (LPL) fractions in AhrdCAIR mice had the higher levels of GFP expression, indicating Ahr transcription, measured by flow cytometry in CD8+ T cells (Figures 1A and 1B). Examination of the different CD8+ T cells displaying markers of effector or memory subsets in the SI LPL (Figure S1A) showed that CD127−KLRG1+ short-lived effector CD8+ T cells (TSLE) had low Ahr expression at a level similar to Tnaive; however, CD127−KLRG1− early effector CD8+ T cells (TEE) and CD127+KLRG1− memory precursor effector CD8+ T cells (TMPE) had higher Ahr expression (Figure 1C). CD44+CD62L+ central memory CD8+ T cells (TCM) had a low level of Ahr expression, while CD44+CD62L− effector CD8+ T cells (TEM) showed higher Ahr expression (Figure 1D). Furthermore, CD69+CD103+ CD8+ T cells (TRM) exhibited the highest level of Ahr expression (Figure 1D). Of note, TRM in the SI LPL and IEL had similar levels of Ahr expression (Figure S1B). However, in contrast to the LPL (Figure S1A), CD8+ T cells in IEL overwhelmingly consisted of the CD69+CD103+ TRM population (>90%) (Figure S1G), consistent with higher expression of Ahr in IEL than in LPL (Figure 1B). Furthermore, we performed analysis of publicly available RNA-seq data of CD8+ T cell subsets following herpes simplex virus (HSV) infection in various tissues (GSE70813). Consistent with our data, the highest expression level of Ahr was detected in TRM (CD69+CD103+), compared with those in TCM (CD44+CD62L+), and TEM (CD44+CD62L−) (Figure S1C). Together, these data suggest in CD8+ T cells Ahr is dynamically expressed along the TRM differentiation pathway in the gut.
Ahr regulates intestinal CD8+ T cell compartment in a cell-intrinsic manner
To further investigate whether Ahr plays a role in the regulation of CD8+ T cell differentiation, we performed a bone marrow chimera experiment. Bone marrow from Ahr+/+ CD45.1/.2 and Ahr−/− CD45.2/.2 donor mice was transferred to half-lethally irradiated CD45.1/.1 mice and allowed to reconstitute for 2 months before flow cytometry analysis of spleen, SI LPL, and IEL (Figure S2A). The percentage of Ahr−/−-derived cells was reduced in the SI IEL with no difference in the LPL or spleen (Figure S2B). This reduction in total lymphocytes was evidently caused by reduced CD8+ T cells, while CD4+ T cells showed no difference (Figures S2C and S2D), suggesting a role of Ahr in regulating CD8+ T cells in SI IEL. However, there was no difference in the protein level of Ki67 (Figure S2E) or annexin V (Figure S2F) expression in IEL Ahr−/− CD8+ T cells examined directly ex vivo, suggesting that the observed decrease in these cells might be due to other mechanisms other than compromised cell proliferation or survival.
To further investigate the cell-intrinsic role of Ahr, we developed a genetic model (Ahrf/fCd8cre) to ablate Ahr expression specifically in CD8+ T cells but not in CD4+ T cells by Cre recombinase transgene under the control of the E8I CD8 enhancer region and the Cd8α promoter.37 Ahrf/fCd8cre mice displayed efficient and specific deletion of Ahr in CD8β+ but not in CD8β− cells (Figures S1D and S1E). Based on examination of different CD8+ T cell subsets in the LPL, we observed TSLE (CD127−KLRG1+) were increased in the Ahrf/fCd8cre mice (Figure S1F, S2G, and S2H) and TRM (CD69+CD103+) cells were modestly but significantly reduced in the SI IEL (Figures S1G, S2I, and S2J). These data suggest that although Ahr is most likely not a driver that directs the development of TRM cells and/or their maintenance, it plays a cell-intrinsic role in promoting TRM compartment at least at the steady state in the gut.
Ahr suppresses TCM while promoting TRM gene signature in intestinal CD8+ T cells
To determine the role of Ahr in regulation of SI epithelial resident CD8+ T cell compartment, we performed gene profiling analysis of CD8+ T cells from the IEL of Ahrf/f and Ahrf/fCd8cre mice via RNA-seq. Principal component analysis and hierarchical clustering showed distinct separation between different genotypes (Figure S3A). 160 genes were differentially expressed (fragments per kilobase million (FPKM) ≥ 1, fold change ≥ 1.5, q ≤ 0.05) when comparing Ahrf/f vs. Ahrf/fCd8cre mice with roughly equal upregulated (57%) and downregulated (43%) genes, with many being a regulator of or associated with CD8+ T cell memory subset fate (Figure 2A and S3B). Gene set enrichment analysis (GSEA) demonstrated a role for Ahr in promoting the expression of TRM-associated genes while suppressing the expression of TCM-associated genes (Figure 2B).28 Notably, the magnitude in which the circulating core gene signature was enriched was greater than the enrichment for the resident core gene signature (Figures 2B and 2C). In the absence of Ahr, multiple transcription factors key to the regulation of CD8+ T cell memory development displayed a selectively differential expression pattern. The TRM-promoting factors Prdm1 and Hic130,38,39 were downregulated, while the TCM-promoting factors Eomes and Klf240,41 showed a trend of increase with no difference in Runx3, Zfp683 (Hobbit), and Tbx21 (T-bet) (Figure 2D). Consistently, at the protein level, in the CD69+CD103+ IEL resident CD8+ T cells of Ahrf/fCd8cre mice, Blimp-1 (encoded by Prdm1) was decreased (Figure S3C), while Eomes was increased (Figure S3D) compared with littermate Ahrf/f control mice. Together, these transcriptomic changes are consistent with the observed TRM defect and enhancement of TCM cells in the gut of Ahr-deficient mice, suggesting a possibility that Ahr plays a role in CD8+ T cell fate decision.
TGFβ and IL-33 have been previously described to enhance in vitro CD69+CD103+ TRM-like cell differentiation.42,43 Of note, Ahr activation with 6-formylindolo(3.2b) carbazole (FICZ), confirmed by expression of the Ahr direct target gene Ahrr (Figure 2I), resulted in a significant enhancement of in vitro TRM-like differentiation and reduced differentiation of the CD44+CD62L+ TCM-like cells. These effects were abrogated in Ahr−/− and Ahr+/+ CH223191-treated (Ahr antagonist) CD8+ T cells (Figures 2J, 2K, S4B, and S4C), consistent with a role for Ahr in regulating TRM and TCM differentiation.
Granzymes, especially granzyme B, a well-described effector molecule secreted by cytotoxic lymphocytes to induce apoptosis in target cells,44 were downregulated at the mRNA level in Ahr-deficient CD8+ cells, while other CD8+ T cell effector molecules including Tnf, Ifng, Prf1, and Ccl4, had no change (Figure 2E), suggesting a selective regulation of CD8+ T cell function in the intestine. Consistently, granzyme B, at the protein level on a per cell basis, was also reduced in Ahr-deficient IEL resident CD8+ T cells (Figures 2F and 2G) as well as in the bone marrow chimera model (Figure S4A).
Ahr directly binds DNA to regulate key TRM genes
To elucidate the direct gene targets of Ahr in CD8+ T cells, we performed chromatin immunoprecipitation followed by sequencing (ChIP-seq) of Ahr using in vitro TRM-like cells. 10,181 unique Ahr binding locations (peaks) were identified with the majority being located in the intron, intergenic, or promoter regions of genes (Figure 3A). Ahr binding at the promoter was enriched given that the relative abundance of promoter regions in the genome is estimated to be about 1%,45 and 30% of Ahr peaks were located at the promoter (Figure 3A). Motif enrichment analysis (Figure 3B) showed the canonical Ahr:Arnt binding motif as the top hit. Other transcription factor motifs such as Runx2, ETS, Batf, IRF, and Stat5 were also found to be enriched, suggesting that Ahr might work together with these factors in a complex to bind DNA and regulate target gene transcription. Integration of the ChIP-seq and RNA-seq data by the software package Binding and Expression Target Analysis (BETA)46 showed that Ahr functioned equally as a transcriptional activator and repressor (Figure 3C). This is different than in other cell types; for example, in ILC2s, Ahr mainly functions as a suppressor,47 suggesting Ahr acts in a cell type-specific manner to regulate the expression of target genes. BETA direct target prediction identified Gzmb together with other TRM-associated genes such as Itgae, Prdm1, and Hic1 as top Ahr-activated genes (Figure 3D), while TCM-characteristic genes such as Klf2, Eomes, S1pr5, and Klrg1 were identified as Ahr-repressed genes (Figure 3D).
To determine whether Ahr DNA binding activity is required for in vitro TRM-like differentiation, Ahr-deficient naive CD8+ T cells were transduced with empty vector (MIG-EV), wild-type Ahr (MIG-Ahr), Ahr DNA binding domain single amino acid mutant (MIG-Y9A),48 or Ahr DNA binding domain deletion mutant (MIG-DbHLH)49 and then differentiated to TRM-like cells. Compared with MIG-EV control, MIG-Ahr transduction increased CD69+CD103+ in vitro TRM-like differentiation, which was further enhanced by Ahr ligand FICZ treatment (Figure 3E) Both Ahr DNA binding mutants failed to upregulate Ahrr, an Ahr direct target gene50,51 (Figure 3F), and exhibited markedly impaired CD69+CD103+ CD8+ T cell induction in TRM-like skewing conditions (Figure 3E). These data suggest that Ahr acts in a DNA-binding-dependent manner to promote in vitro TRM-like differentiation.
Given that Blimp1 (encoded by Prdm1), a well-known factor in promoting tissue residency,30 was identified as an Ahr direct target (Figure 3D), we further transduced Ahr−/− CD8+ T cells with hCD2-Blimp1. Notably, forced expression of Blimp1 resulted in a marked enhancement of in vitro TRM-like differentiation similar to the MIG-Ahr transduced cells (Figures 3G and S5A). These data showed that restoration of Blimp1 levels was sufficient to rescue the observed in vitro TRM-like differentiation defect in Ahr-deficient CD8+ T cells, suggesting that Blimp1 is one of the major downstream targets directly regulated by Ahr during TRM differentiation. It is possible that the in vitro over-expression assay would not faithfully recapitulate the in vivo microenvironment and thus may not confer the whole TRM gene expression program. Consistent with the regulation of CD69 and CD103 expression by Ahr, we analyzed Ahr ChIP-seq data and showed that Ahr directly bound to both the Cd69 and Itgae (CD103) gene loci (Figures S5B and S5C).
Ahr is a cell-intrinsic promoter of intestinal CD8 TRM during infection
The reduction of CD69+CD103+ cells in the gut of Ahr-deficient mice under the steady state albeit significant is a modest reduction. Additionally, the results from the in vitro TRM-like differentiation could be interpreted as Ahr regulating the expression of CD69 and CD103, not the TRM population as a whole. Indeed, Ahr ChIP-seq analysis showed that Ahr directly bound to both the Cd69 and Itgae (CD103) gene loci (Figures S5B and S5C). Therefore, we sought to determine the role of Ahr in CD8+ T cell responses during infection to further validate these findings. To this end, we utilized a TRM-dependent oral Listeria monocytogenes-ovalbumin (L.m.-OVA) infection model.3 The reasons for utilization of this model are 2-fold. It is an intestine-specific infection model that recapitulates human infection, allowing us to corroborate our findings on the role of Ahr in CD8+ T cells under the steady state of the gut; in addition, in combination with OVA-specific transgenic T cells (OTI) it allows for tracking mucosal CD8+ T cell responses in an antigen-specific manner. Host CD45.1/.1 mice were co-transferred with CD45.1/.2 Ahr+/+ and CD45.2/.2 Ahr−/− naive OTI CD8+ T cells before infection (Figure 4A). In the SI IEL, CD45.2/.2 Ahr−/− OTI cells were significantly and gradually reduced (Figure 4B). At an effector time point (day 9 post infection), while there was no difference in their frequency in the IEL, Ahr−/− OTI cells already exhibited an imbalanced memory precursor response. Specifically, CD127+ memory precursor populations were reduced in the Ahr−/− OTI cells with a concomitant increase of CD127−KLRG1+ TSLE (Figure 4C). Immature CD127−KLRG1− TEE was also increased in the Ahr−/− OTI cells. The significance of the CD127/KLRG1 memory precursor vs. short-lived effector paradigm in the intestine is not well described. However, during the early stage of infection, CD69+CD103+ cells could be identified and are suggested to give rise to TRM in the memory phase.3 Indeed, at this effector time point, Ahr−/− cells had already shown a reduction in the CD69+CD103+ population with an increase in CD69−CD103− cells (Figure 4D), consistent with defective CD8+ TRM differentiation due to Ahr deficiency.
In the spleen, we observed a similar frequency of OTI CD8+ T cells with or without Ahr; however, in MLNs there was an increase in Ahr-deficient OTI CD8+ T cells (Figures S6A and S6B), suggesting the priming of these cells is not impaired. Phenotypic analysis showed that in the absence of Ahr, TCM-like (CD44+CD62L+) and TMPE (CD127+KLRG1−) cells were increased in both the spleen and MLNs (Figures S6C–S6J), consistent with the notion that Ahr might function as a TCM suppressor. Of note, in agreement with a previous report,3 we observed a higher frequency of TMPE in the spleen in oral L.m.-OVA infection than those typically observed during systemic infection models. Additionally, while CD69+CD103+ cells were not present in the mesenteric lymph nodes (MLNs) at this time point, Ahr deficiency led to a decreased frequency of CD69+CD103− cells that might be poised to become TRM26 (Figures S6K and S6L), suggesting that Ahr acts early to promote TRM.
To further determine Ahr-mediated regulation of CD8+ T cell differentiation in the IEL at the early time point of infection, we performed RNA-seq analysis of Ahr+/+ and Ahr−/− OTI CD8+ T cells isolated from day 9 post L.m.-OVA infected mice. Consistent with our hypothesis that Ahr regulates the differentiation of TRM, we observed, in the absence of Ahr, a suppression of TRM signature gene expression (Figure S7A) with multiple genes overlapping with the steady state RNA-seq analysis (highlighted in red) including the Ahr direct target Tiparp and the TRM master regulator Prdm1 (Figure S7B). Comparison of the two datasets, by principal component analysis, showed that upon addition of the day 9 post-infection data, the steady-state samples were no longer separated based on genotype, but for the infection dataset, Ahr+/+ samples were clustered together and distinct from the Ahr−/− samples, suggesting that infection magnifies the gene expression changes between Ahr−/− and Ahr+/+ CD8+ T cells that were observed under the steady state (Figure S7C). Furthermore, visualization of the transcriptome-wide fold change showed both in number and magnitude greater differential gene expression in the infection dataset compared with steady state (Figure S7D), suggesting that upon challenge with an infectious agent the impact of Ahr deficiency on CD8+ transcriptome is greater than that under the steady state. Consistently, the magnitude of the Residency Core Gene Signature enrichment was larger, and the number of differential genes was more compared with the steady state dataset (Figures 2B and 2C vs. S7A and S7B). Collectively, these data suggested that Ahr might play a more prominent role in shaping TRM gene program during infection than it does at steady-state “physiological inflammation” in the gut.
At the memory phase of infection, bona fide TRM is the sole CD8+ T cell subset that remains in the IEL by day 34 post infection.3 Given the reduction of TRM precursor cells observed at day 9, we hypothesized that Ahr-deficient OTI cells would be reduced at this memory time point. Indeed, in the IEL, CD45.2/.2 Ahr−/− OTI cells were significantly reduced at day 34 (Figure 4A). Additionally, CD45.2/.2 Ahr−/− cells produced less granzyme B on a per cell basis compared with CD45.1/.2 Ahr+/+ OTI cells (Figures 4E and 4F). Also, at this time point and consistent with the decreased expression of Prdm1 in Ahr-deficient CD8+ T cells analyzed by RNA-seq, IEL OTI cells of Ahr−/− origin exhibited lower levels of Blimp1 staining compared with Ahr+/+ cells (Figures S7E and S7F). Collectively these data suggest a critical role of Ahr in promoting TRM cells and their function during infection potentially through regulation of Blimp1 expression.
To determine if the observed reduction of Ahr−/− CD8+ TRM cells also was present after a secondary recall challenge, following primary infection, mice were re-infected with L.m.-OVA, and then 3 days post re-infection, mice were sacrificed, and analysis of OTI-specific T cell responses was performed (Figure 4A). Consistent with results from the primary infection, in the IEL, CD45.2/.2 Ahr−/− OTI cells were significantly reduced (Figures 4G and 4H) and produced less granzyme B (Figures 4I and 4J). However, in the spleen there was no difference in granzyme B production (Figures S7G and S7H), while Ahr−/− OTI cells from the MLNs showed a trend of decreased granzyme B (Figures S7I and S7J), suggesting that Ahr regulation of CD8+ T cell function might be specific to the gut. Together, these results demonstrate that Ahr is a cell-intrinsic promoter of long-lived, functional TRM cells during intestinal infection.
Single-cell RNA-seq identifies polyfunctional CD8+ T cell population dependent on Ahr
CD8+ tumor-infiltrating lymphocytes (TILs) displaying a TRM-like phenotype have been well described to be critical to the orchestration of potent protective anti-tumor response.52,53 Therefore, we next investigated the role of Ahr in CD8+ T cells in tumor immunity. Previous literature suggests that Ahr can function as either a tumor promoter or suppressor dependent of the context.54,55 However, these studies did not address the cell-intrinsic role of Ahr in the tumor, in which perturbation of Ahr in different cell types such as cancer cells or immune cells would inevitably have different outcomes of tumorigenesis. To elucidate the role of Ahr in tumor immunity, B16F10 melanoma cells were injected subcutaneously into Ahr−/− mice with Ahr germline deletion and into their littermate wild-type Ahr+/+ mice. CD45+ TILs were purified and subjected to high-throughput single-cell RNA-seq using the 10X Genomics platform. 28 different clusters were identified in both groups with no marked differences between groups. (Figures S8A–S8C).
Given the crucial role of CD8+ T cells in anti-tumor immunity,28,53 we decided to focus on the impact of Ahr on CD8+ T cells in the tumor microenvironment. Particularly of interest are CD8+ TILs labeled “polyfunctional” that simultaneously produce multiple effector molecules including cytokines, chemokines, and cytotoxic granules. Subsetting on the CD8+ T cell clusters and re-performing dimensionality reduction, clustering, marker gene finding, and visualization resulted in six unique clusters with visual differences between Ahr+/+ and Ahr−/− (Figures 5A and S8D). Ahr-deficient CD8+ T cells were decreased in the polyfunctional and activated (Ifit1+ and Il1b+) subsets while having an increase in the cluster with a circulating memory-like phenotype (Figure 5A right). Pseudotime analysis by Monocle3 showed that Ahr-deficient CD8+ T cells were less advanced in the pseudotime differentiation trajectory (Figures 5B and 5C), consistent with a role for Ahr in the regulation of CD8+ T cell differentiation. Differential gene expression analysis showed a reduction of genes critical for CD8+ T effector function (i.e., Gzmb, Prf1, Ifng, Ccl4, and Xcl1), residency-associated membrane receptors (i.e., Icos and Cd69), and transcriptional modulators (i.e., Smad7, Runx2, and Hif1a) (Figure 5D). Collectively, these results indicate that Ahr is critical to the development of highly activated and polyfunctional CD8+ TILs.
To understand the CD8+ T cell-intrinsic role of Ahr in tumor immunity, Ahrf/fCd8cre mice were challenged with the aforementioned B16F10 melanoma model and exhibited significantly enhanced tumor growth (Figure 5E) and burden at endpoint (Figure 5F) compared with controls. While there were no differences in their frequency (Figure S8E), CD8+ TILs of Ahrf/fCd8cre mice were less polyfunctional and produced less granzyme B, IFNγ, and TNF, on a per cell basis. Triple-positive cells (granzyme B+IFNγ+TNF+) were reduced in the Ahrf/fCd8cre mice, while triple-negative cells (granzyme B−IFNγ−TNF−) were increased in the Ahrf/fCd8cre mice (Figures 5G, 5H, S8E, and S8F). Given the low immunogenicity of the B16F10 model, we sought to validate these findings using the MC38 murine colon carcinoma model. Consistently, Ahrf/fCd8cre mice exhibited significantly enhanced tumor growth (Figure S9A) and burden at endpoint (Figure S9B) compared with littermate controls. Furthermore, CD8+ TILs of Ahrf/fCd8cre mice produced less granzyme B, IFNγ, and TNF on a per cell basis (Figures S9C and S9D) and were less polyfunctional (Figure S9E) in the MC38 tumor model as well. Collectively, these results suggested that Ahr promotes polyfunctional CD8+ TILs in a cell-intrinsic manner.
AHR signaling promotes TRM differentiation and function in human CD8+ T cells
To determine the role of AHR in human CD8+ T cells, we next examined the expression of AHR in human intestine resident CD8+ T cells. Compared with blood circulating CD45RA−CD45RO+ CD8+ T cells, virtually all IEL resident counterparts were CD69+CD103+ (Figure 6A). As expected, human IEL resident CD8+ T cells expressed less T-bet,56 but more AHR (Figures 6B and 6C), consistent with the observation of enrichment of Ahr expression in mouse IEL CD8+ T cells. Next, we performed human in vitro TRM-like differentiation using TGFβ and IL-33. Treatment with CH223191, a specific Ahr antagonist,51 caused a reduction in the frequency of CD103+ cells, whereas FICZ enhanced this population (Figure 6D). Although FICZ or CH223191 treatment did not significantly impact AHR expression, TGFβ and IL-33 treatment enhanced AHR expression in human CD8+ T cells (Figure 6E). As expected, FICZ treatment increased expression of the AHR target gene AHR, while CH223191 suppressed its expression. Notably, FICZ also significantly augmented the expression of the effector gene GZMB (Figures 6F and 6G). Together, these results demonstrate an evolutionarily conserved function of AHR in human CD8+ T cells.
DISCUSSION
Proper regulation of CD8+ T cell memory development is critical to long-term protection against morbid infection. Multiple transcription factors have been suggested to contribute to the regulation of this process; however, data describing their cell-intrinsic effects are lacking. A role for Ahr supporting TRM has been previously described.57 Ahr expression was shown to be higher in skin compared with spleen CD8+ T cells, and Ahr functions to facilitate TRM persistence in the epidermis.57 Our study provided mechanistic insights into how Ahr regulates the transcriptional programs of CD8+ T cells and elucidated a role for Ahr in early TRM differentiation, specifically in a gut-specific infection model.
Other studies, using an influenza infection model, demonstrated that early-life Ahr activation via TCDD impairs the priming of virus-specific CTLs; however, this phenotype is mediated by regulation of DNA methylation and found to be cell extrinsic.58,59 Of additional importance, the cellular toxicity and long half-life of TCDD could lead to non-physiological activation of Ahr, thus complicating data interpretation. Given that Ahr is expressed in various cell types and plays a complex role, we ablated Ahr expression specifically in CD8+ T cells. We noted that differences in TRM under the steady state between Ahr-deficient mice and their littermate controls were modest, presumably due to the lack of antigen-specific pathogen exposure and non-competitive environment. Although our data suggest that Ahr plays a cell-intrinsic role in CD8+ T cell differentiation and function, we opted to test this hypothesis in the context of gut infection as well. In the intestine, on day 9 post oral infection, we showed that Ahr-deficient CD8+ T cells exhibited a reduction of TRM precursors with a concomitant increase in TSLE and TCM-like cells compared with wild-type counterparts. This decrease in precursors led to a marked reduction in the IEL resident TRM cell compartment at the memory time point of day 34 post infection. Of note, for the gut infection model Ahr-deficient OTI CD8+ T cells responded to infection in the presence of an equal number of wild-type OTI CD8+ T cells. This allows for a well-controlled experiment because both are exposed to the same environment and stimuli; however, the magnitude of the observed phenotype could be due to competition. Furthermore, we showed that Ahr functions to promote in vitro TRM-like cell differentiation while suppressing the TCM phenotype. Collectively, these data prompted us to conclude that differentiation at least is one of the key mechanisms underlying the defective CD8+ T cell memory development that occurs in the absence of Ahr.
A previous report has shown Ahr is a critical regulator of monocyte differentiation acting to promote dendritic cell differentiation while suppressing macrophage differentiation.60 In the current study, we demonstrate that in CD8+ T cells, Ahr acts as a cell fate decision regulator to suppress the differentiation of TCM while promoting TRM. Blimp1 is a well-described positive regulator of TRM differentiation and function.29 We showed that Prdm1 (Blimp1) is one of the top Ahr-regulated direct target genes in CD8+ T cells, consistent with the role of Ahr in promoting Prdm1 expression in monocytes.60 We further showed that forced expression of Blimp1 was able to rescue in vitro TRM-like differentiation defect in the absence of Ahr, suggesting that Blimp1 acts downstream of Ahr to regulate CD8+ T cell memory development. It is important to note that Ahr−/− CD8+ T cells were still able to differentiate into CD69+CD103+ cells in vitro and in vivo; therefore, Ahr is likely acting as a promoter rather than a main driver of TRM and in vitro TRM-like differentiation.
Recent publications describe that Ahr promotes CD8+ T cell exhaustion in tumor immunity.61,62 However, activated T cells as well as tissue-resident memory cells express many of the exhaustion-associated molecules (PD-1, Tim-3, CTLA-4, etc.).28,56,63 It is also important to keep in mind that interpretation of the role of Ahr in tumor immunity with ligand treatment has to consider both cell-autonomous and non-autonomous effects, given the cell-specific role of Ahr in different immune cell types that infiltrate the tumor microenvironment, and in the tumor cells as well. It has been reported that when TRM responses are diminished, polyfunctional CD8+ TILs are also reduced,64,65 consistent with our findings. These polyfunctional tumor-resident memory-like cells have been positively associated with productive anti-tumor immunity.66–68
Activation of the Ahr pathway in CD8+ T cells may be of therapeutic interest in the context of infection or cancer immunotherapy when a long-lived tissue-resident memory response is of utmost importance.
Limitations of the study
Our current study represents a serious effort to delineate the cell-intrinsic role of Ahr in CD8+ T cells, specifically in the gut. However, future efforts to understand the role of Ahr in CD8+ T cells in other infection models including skin infection with HSV and systemic lymphocytic choriomeningitis virus (LCMV) infection would help extend our understanding of this biology. Although no difference in the proliferation or cell death was observed in the intestinal Ahr-deficient TRM cells ex vivo, whether Ahr also regulates TRM maintenance in the intestine remains to be carefully determined. Future investigation is needed to elucidate the cell-intrinsic role of Ahr in regulation of TRM maintenance, for example by development of CD8-specific inducible deletion of Ahr mouse model to examine TRM and TRM-like cells in various tissues under the steady state and during infection. Additionally, it is possible that Ahr deficiency could lead to decreased TRM due to aberrant re-entering into circulation after having been in the tissue. In the future, more careful analysis of the recirculation of TRM in the context of Ahr deficiency needs to be done through TRM fate mapping mice as previously described.32
STAR★METHODS
RESOURCE AVAILABILITY
Lead contact
Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead contact, Liang Zhou (liangzhou497@ufl.edu).
Materials availability
This study did not generate new unique reagents.
Data and code availability
All data supporting the findings of this study are available within the article and its supplementary information and from the corresponding author upon reasonable request. The accession number of the RNA-Seq, ChIP-Seq, and scRNA-Seq data reported in this paper have been uploaded to GEO at the accession number: GSE220944. This paper does not report original code. Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.
EXPERIMENTAL MODEL AND SUBJECT DETAILS
Human sample collection
Buffy coats from male healthy donors were purchased from Life South Community Blood Centers in accordance with the Institutional Review Boards at the University of Florida (UF IRB #201801563) and PBMCs isolated as described previously.51 Patients (both male and female) diagnosed with an IBD [Crohn disease (CD) and UC] were recruited for this study. Patients were at least 18 years old and recruited during clinical visits to the Inflammatory Bowel and Celiac Disease Program or before a previously scheduled colonoscopy. Tissue biopsies were taken from the colon. The mucosa was assessed based on frequently used scoring systems (Mayo Score for UC and Simple Endoscopic Score for Crohn Disease for CD). Biopsies collected were labeled as “healthy” based on normal vascularity and lack the presence of edema, erythema, friability, erosions, or ulcerations macroscopically. Intestinal tissue biopsies were collected in complete RPMI and then immediately processed to isolate lamina propria lymphocytes (LPLs) and intraepithelial lymphocytes (IELs) as described previously.69
Mice
Mice used in this study were maintained in specific-pathogen-free (SPF) conditions at the University of Florida. All mouse studies were approved by the Institutional Animal Care and Use Committees of the University of Florida. Littermate controls as well as both male and female mice were used for experiments. Mice were used at 8 to 10-week-old age unless otherwise noted. Ahr−/−,70 AhrdCAIR 36, Ahrf/f 51 were published previously. EI8Cre mice, and OTI TCR transgenic mice were purchased from Jackson Laboratory then crossed with Ahrf/f and Ahr−/− mice, respectively.
METHOD DETAILS
Lymphocyte isolation and flow cytometry
The isolation of lymphocytes from spleen, peripheral/mesenteric lymph nodes, small and large intestinal lamina propria, and small intestine intraepithelial lymphocytes were performed as previously described.71 After digestion, cells were further purified from the interphase of 37.5% and 75% Percoll gradient after 20 min spin at 2,500 rpm at room temperature. For flow cytometry analysis, cells were stained using Live and Dead violet viability kit (Invitrogen) or Zombie Aqua fixable viability kit (BioLegend). CD16/32 antibody (Thermo Fisher) was used to block the nonspecific binding followed by surface molecule staining on ice for 30 min. Cells were fixed and permeabilized with Foxp3 staining buffer Kit (eBioscience) for transcription factor staining. For cytokine staining, cells were stimulated with 50 ng/mL PMA and 500 ng/mL ionomycin for 3 h and Brefeldin A (2 μg/mL) was added 2 h before cells were collected. Sample acquisition was performed on BD FACSCantoII or Cytek Aurora flow cytometer and analyzed with FlowJo software (version 10.2).
Adoptive transfers and infection
The spleens of CD45.1/.2 Ahr+/+ OT-I and CD45.2/.2 Ahr−/− OT-I were isolated and single-cell suspensions were generated by mechanical disruption. Naive CD8 (Tcrb+CD8a+CD44lo) OT-I T cells were first enriched using mouse naive CD8+ T cell isolation kit (Stemcell Technologies) then further purified using a Sony sorter SH800. Post-sort purity was routinely analyzed and higher than 97%. Purified cells were then mixed in a ratio of 1:1 and 5 × 104 cells were co-transferred 1 day before infection. Mice were infected orally by feeding 2.5 × 109 (primary infection) colony-forming units (CFUs) of L.m.-OVA InlAM (kindly provided by B. Sheridan, Stony Brook University) as described previously.3 At the indicated time points after infection, mice were sacrificed, and tissues were collected for analysis of OT-I T cell responses. For experiments investigating the recall response, following primary infection (>30 day later) mice were re-infected with 2.5 × 1010 CFU of L.m.-OVA InlAM. Then, 3 days later mice were sacrificed, and tissues were processed for analysis of OT-I T cell responses.
In vitro TRM differentiation
CD8 TRM cells were differentiated in vitro as described previously.42 Briefly, naive CD8+ T cells were purified from splenocytes using mouse naive CD8+ T cell isolation kit (Stemcell Technologies) then cultured in RPMI-1640 medium (plus β-mercaptoethanol) supplemented with 10% fetal bovine serum, 1% L-glutamine, 1% penicillin-streptomycin (cRPMI). 100 ng/mL IL-2 and anti-CD3/CD28 coated Dynabeads (Thermo Fisher) at 1:1 cell to bead ratio were added and cells incubated for 48 h. Then media volume was doubled, each well split into a second well, and 5 ng/mL TGF-β and 100 ng/mL IL-33 were added and incubated for another 48 h before harvest for flow cytometry and RT-qPCR analysis.
For human cells, naive CD8+ T cells were purified from PBMCs using human naive CD8+ T cell isolation kit (Stemcell Technologies) then cultured in cRPMI and treated as above with the human equivalent Dynabeads and cytokines. FICZ and CH223191 were added at a concentration of 200 nM and 1μM, respectively, as indicated in the text.
Quantitative PCR
Total RNA was isolated with Trizol reagent (Invitrogen). cDNA was synthesized by GoScript™ Reverse Transcription Kit (Promega). Real-time PCR was performed using SYBR Green (Biorad) and different primer sets (Table S1). Reactions were run using the CFX Connect Real-Time PCR Detection System (Biorad). Each specific gene expression was normalized to β-actin expression.
RNA-seq and ChIP-Seq assay and analyses
For RNA-Seq analyses of IEL resident CD8αβ and CD8αα T cells, 2×103 sorted CD8αβ and CD8αα T cells from the small intestine IEL of control or Ahrf/fCd8cre littermate mice were used. RNA was isolated by RNAeasy Micro Kit. cDNA generation was performed with SMART-Seq® HT Kit (Takara). Sequencing libraries were generated with Nextera® XT DNA Library Preparation Kit (Illumina). Libraries were sequenced on an Illumina HiSeq 4000 instrument to produce 50 bp single-end reads. The read mapping and mRNA quantification were performed as previously described.72 Briefly, FastQC was used to ensure high per-base sequence quality of reads. Sequenced reads were mapped and raw count values quantified with STAR73 to the Mus musculus genome (GRCm38/mm10 assembly). RSEM74 was used to quantify mRNA expression levels, FPKM aligned reads. Differentially expressed genes (max FPKM ≥1, fold change ≥1.5, q-value ≤0.05) were identified by DESeq275 analysis. Gene Set Enrichment Analysis (GSEA)76,77 were performed using the circulating and resident gene signatures developed previously.28 Log2 transformed FPKM values were used for principal component analysis in R78 with the prcomp function and then visualized using the rgl79 package. Heatmaps were created using the R package pheatmap.80
For ChIP-Seq analyses of in vitro TRM, cells were differentiated from naive CD8+ T cells as described above. Cells were then treated with FICZ (200 nM) for 4 h before harvest. Afterward, cells were collected and cross-linked with 1% formaldehyde for 15 min. Chromatin was sheared by sonication with Bioruptor Pico (30″ on and 30″ off for 25 cycles) and immunoprecipitated with anti-Ahr antibody (Enzo Life Science) using iDeal ChIP-Seq Kit for transcription factors or True MicroChIP Kit (Diagenode). Eluted DNA was used to generate an indexed library according to the manual of NEBNext Ultra II DNA Library Prep Kit for Illumina (NEB). The library was cleaned up with 1.2x SPRIselect beads (Beckman Coulter) before sequencing on an Illumina HiSeq 4000 instrument to produce 50 bp single-end reads. The read mapping and ChIP-Seq analysis were performed as previously described.51 Briefly, FastQC was used to ensure high per-base sequence quality of reads. Then ChIP-Seq reads were mapped to the mouse genome (GRCm38/mm10 assembly) with bowtie2 (v2.3.3)81 and further filtered using samtools (v1.7).82 The uniquely aligned reads were used to generate bedgraph files (scaled to 10 million reads) using bedtools (v2.25.0)83 and then were uploaded to UCSC genome browser for visualization. ChIP-Seq peak finding, Motif enrichment analysis, and peak annotation were performed using Homer84 with default parameters.
Retroviral transduction
Human embryonic kidney (HEK) 293T cells were transfected with retroviral plasmids and the packaging plasmid 10A1 using polyethyleneimine (PEI). Viral supernatant was collected after transfection. CD8+ T cells were isolated and cultured as described above. After 24 h of culture, retrovirus-containing supernatants supplemented with polybrene (8 μg/mL, Sigma-Aldrich) were added to the cells followed by centrifugation at 2500 rpm for 2 h at 32°C on days 1 and 2. The cells were further cultured under in vitro TRM-like differentiation conditions before harvest as indicated in the text.
Plasmids
cDNA of mouse Ahr (1–805 amino acids) was cloned into MIG with hemagglutinin (HA) at the N terminus. For the Ahr Y9A (1–805 amino acids, Y9A), AhrΔbHLH (1–120 amino acids, were deleted) and subcloned into MIG with HA tag. Blimp1-hCD2 plasmid was kindly provided by Dr. Weishan Huang (Louisiana State University).
Tumor model
For the melanoma and MC38 tumor models, mice were injected in the right flank subcutaneously with B16-F10 or MC38 (5 × 105) cells then tumor growth was monitored every other day with a digital caliper. At endpoint, tumors were excised and cut with scissors into <2mm pieces then incubated with collagenase and DNase I for 30 min at 37°C in a shaking incubator. Samples were passed through a 100-μm filter, tumor infiltrating lymphocytes (TILs) were isolated by interphase collection after a 40% and 80% Percoll gradient centrifugation and then analyzed via flow cytometry as described above.
Single cell RNA-Seq analyses
Live CD45+ total TILs were isolated and purified by FACS sorting from a total of 4 mice (2 Ahr+/+ and 2 Ahr−/−) before being loaded on the chromium controller aiming for a recovery of 10,000 cells. Single Cell 30 reagent kit v3.1 was used for reverse transcription, cDNA amplification, and library construction of gene expression libraries (10x Genomics) according to the manufacturer. For primary scRNA-Seq analysis: alignment, quantification, and quality control were performed using the Cell Ranger Software and default parameters. After quality control and removal of dead cells, doublets, and contaminating melanoma cells (Pmel mRNA count >0) a total of 9726 cells remained for downstream analysis. Normalization, cell clustering (resolution = 2), dimensionality reduction, differential expression, visualization, and pseudotime analysis were performed using the R packages Seurat85 and Monocle.86 The 3000 most variable features identified with the variance-stabilizing transformation (vst)-method were used for principal component analysis (PCA). Upon inspection of elbowplot and jackstraw plot, the first 20 principal components (PCs) were used for further analysis. Cell type annotation was performed manually by assessment of cluster marker genes.
QUANTIFICATION AND STATISTICAL ANALYSIS
All data are represented as mean ± SEM and have at least n = 3 per group from at least 2 independent experiments (refer to figure legend to detailed information). Unless otherwise noted, statistical analysis was performed with the unpaired Student’s t-test or one-way ANOVA with Dunnett’s correction for multiple comparisons. For analysis of tumor growth kinetics, linear regression was performed to test if the slopes are significantly different between groups. Statistical analyses were run using GraphPad Prism 8 software package. p values were indicated with asterisks (*p % 0.05; **p % 0.01; ***p % 0.001; ****p % 0.0001).
Supplementary Material
KEY RESOURCES TABLE.
REAGENT or RESOURCE | SOURCE | IDENTIFIER |
---|---|---|
| ||
Antibodies | ||
| ||
Anti-mouse CD45.2 – FITC (clone 104) | TONBO biosciences | Cat# 35-0454-U500; RRID: AB_2621692 |
Anti-mouse CD45.1 – APC/Cyanine7 (clone A20) | BioLegend | Cat# 110716; RRID: AB_313505 |
Anti-mouse TCRβ – Brilliant Violet 650 (clone H57-597) | BioLegend | Cat# 109251; RRID: AB_2810348 |
Anti-mouse TCRβ – eFluor 450 (clone H57-597) | Invitrogen | Cat# 48-5961-82; RRID: AB_11039532 |
Anti-mouse CD3e – FITC (clone 145-2C11) | TONBO biosciences | Cat# 35-0031-U500; RRID: AB_2621659 |
Anti-mouse CD3 – APC-eFluor 780 (clone 17A2) | Invitrogen | Cat# 47-0032-82; RRID: AB_1272217 |
Anti-mouse CD8b – FITC (clone YTS156.7.7) | BioLegend | Cat# 126606; RRID: AB_961295 |
Anti-mouse CD8b – Brilliant Violet 711 (clone YTS156.7.7) | BioLegend | Cat# 126633; RRID: AB_2800622 |
Anti-mouse CD8a – APC/Cyanine7 (clone 53-6.7) | BioLegend | Cat# 100714; RRID: AB_312753 |
Anti-mouse CD8a – Brilliant Violet 605 (clone 53–6.7) | BioLegend | Cat# 100744; RRID: AB_2562609 |
Anti-mouse/human CD44 – Brilliant Violet 421 (clone IM7) | BioLegend | Cat# 103040; RRID: AB_2616903 |
Anti-mouse CD62L – PE-Cyanine7 (clone MEL-14) | Invitrogen | Cat# 25-0621-82; RRID: AB_469633 |
Anti-mouse CD127 – PE/Dazzle 594 (clone A7R34) | BioLegend | Cat# 135032; RRID: AB_2564217 |
Anti-mouse KLRG1 – PerCP-eFluor 710 (clone 2F1) | Invitrogen | Cat# 46-5893-82; RRID: AB_10670282 |
Anti-mouse CD69 – Brilliant Violet 711 (clone H1.2F3) | BioLegend | Cat# 104537; RRID: AB_2566120 |
Anti-mouse CD69 – PE (clone H1.2F3) | Invitrogen | Cat# 12-0691-82; RRID: AB_465732 |
Anti-mouse CD103 – Brilliant Violet 785 (clone 2E7) | BioLegend | Cat# 121439; RRID: AB_2800588 |
Anti-mouse CD103 – APC (clone 2E7) | Invitrogen | Cat# 17-1031-82; RRID: AB_1106992 |
Anti-human/mouse Granzyme B – Pacific Blue (clone GB11) | BioLegend | Cat# 515408; RRID: AB_2562196 |
Anti-human/mouse Granzyme B – Alexa Fluor 700 (clone QA16A02) | BioLegend | Cat# 372222; RRID: AB_2728389 |
Anti-mouse IFN gamma – PerCP-Cyanine5.5 (clone XMG1.2) | Invitrogen | Cat# 45-7311-82; RRID: AB_1107020 |
Anti-mouse TNF – PE-Cyanine7 (clone MP6-XT22) | Invitrogen | Cat# 25-7321-82; RRID: AB_11042728 |
Anti-mouse Ki-67 – Brilliant Violet 605 (clone 16A8) | BioLegend | Cat# 652413; RRID: AB_2562664 |
Anti-mouse Ki-67 – Alexa Fluor 647 (clone B56) | BD | Cat# 558615; RRID: AB_647130 |
Anti-mouse Eomes – PE-Cyanine7 (clone Dan11mag) | Invitrogen | Cat# 25-4875-82; RRID: AB_2573454 |
Anti-mouse Ahr – eFluor 660 (clone 4MEJJ) | Invitrogen | Cat# 50-5925-82; RRID: AB_2574255 |
Anti-mouse Ahr – Alexa Fluor 488 (clone 4MEJJ) | Invitrogen | Cat# 53-5925-82; RRID: AB_2574425 |
Anti-mouse Blimp1 – APC (clone 5E7) | BioLegend | Cat# 150007; RRID: AB_2728186 |
Anti-Annexin V – APC | eBioscience | Cat# 17-8007-74 |
Anti-Annexin V – PE/Dazzle 594 | BioLegend | Cat# 640955 |
Anti-human CD3 – FITC (clone SK7) | TONBO biosciences | Cat# 35-0036-T025; RRID: AB_2621661 |
Anti-human CD8 – APC-Cyanine7 (clone SK1) | TONBO biosciences | Cat# 25-0087-T025; RRID: AB_2848136 |
Anti-human CD45RA – PerCP-Cyanine5.5 (clone HI100) | TONBO biosciences | Cat# 65-0458-T100; RRID: AB_2621896 |
Anti-human CD45RO – Brilliant Violet 605 (clone UCHL1) | BioLegend | Cat# 304237; RRID: AB_2562143 |
Anti-human CD197 – Brilliant Violet 711 (clone G043H7) | BioLegend | Cat# 353227; RRID: AB_11219587 |
Anti-human CD127 – PE/Dazzle 594 (clone A019D5) | BioLegend | Cat# 351335; RRID: AB_2563636 |
Anti-human CD69 – Brilliant Violet 785 (clone FN50) | BioLegend | Cat# 310931; RRID: AB_2561370 |
Anti-human CD103 – APC (clone Ber-ACT8) | BioLegend | Cat# 350215; RRID: AB_2563906 |
Anti-human AhR – PE (clone T49-550) | BD | Cat# 565711; RRID: AB_2739336 |
Anti-human T-bet – Brilliant Violet 421 (clone 4B10) | BioLegend | Cat# 644815; RRID: AB_10896427 |
Anti-human CD2 – eFluor 450 (clone RPA-2.10) | Invitrogen | Cat# 48-0029-42; RRID: AB_2574006 |
anti-mouse Ahr (polyclonal) | Enzo Life Science | Cat# BML-SA210–0100; RRID: AB_10540536 |
| ||
Bacterial and virus strains | ||
| ||
L.m.-OVA InlAM | Sheridan et al., 20143 | N/A |
| ||
Biological samples | ||
| ||
Human PBMC isolated from buffy coats | Life South Community Blood Centers | https://www.lifesouth.org/ |
Human colon tissue biopsies | UFHealth Inflammatory Bowel and Celiac Disease Program | https://ufhealth.org/comprehensive-inflammatory-bowel-diseases-program/overview |
| ||
Chemicals, peptides, and recombinant proteins | ||
| ||
Recombinant Murine IL-2 | PeproTech | Cat# 212-12 |
Recombinant Murine IL-33 | PeproTech | Cat# 210-33 |
Recombinant Human TGF-β | PeproTech | Cat# 100-21 |
Recombinant Human IL-2 | PeproTech | Cat# 200-02 |
Recombinant Human IL-33 | PeproTech | Cat# 200-33 |
6-formylindolo(3.2b) carbazole (FICZ) | Sigma | Cat# SML 1489-5MG |
CH223191 | Sigma | C8124-5MG |
| ||
Critical commercial assays | ||
| ||
TRIzol Reagent | Invitrogen | Cat# 15596018 |
iQ SYBR Green Supermix | Biorad | Cat# 1708887 |
Nextera DNA Library Preparation Kit | Illumina | Cat# FC-121-1030 |
NEBNext High-Fidelity 2X PCR Master Mix | NEB | Cat# M0541 |
SMART-Seq HT Kit | Takara | Cat# 634456 |
iDeal ChIP-Seq | Diagenode | Cat# C01010055 |
GoScript Reverse Transcriptase | Promega | Cat# A5003 |
Zombie Aqua Fixable Viability Kit | BioLegend | Cat# 423102 |
Live and Dead Violet Viability Kit | Invitrogen | Cat# L34955 |
Foxp3/Transcription Factor Staining Buffer Set | eBioscience | Cat# 00-5523-00 |
Dynabeads Mouse T-Activator CD3/CD28 | Thermo Fisher | Cat# 11452D |
Dynabeads Human T-Activator CD3/CD28 | Thermo Fisher | Cat# 11131D |
| ||
Deposited data | ||
| ||
RNA-Seq, ChIP-Seq, scRNA-Seq | This paper | GEO: GSE220944 |
| ||
Experimental models: Cell lines | ||
| ||
B16F10 | ATCC | CRL-6475 |
MC38 | Kerafast | ENH204-FP |
| ||
Experimental models: Organisms/strains | ||
| ||
Mouse: CD45.1/1 | The Jackson Laboratory | Cat# 002014 |
Mouse: Ahr−/− | Fernandez-Salguero et al. | N/A |
Mouse: AhrdCAIR | Ye et al., 201736 | N/A |
Mouse: EI8Cre | The Jackson Laboratory | Cat# 008766 |
Mouse: Ahrf/f | The Jackson Laboratory | Cat# 035734 |
Mouse: OTI TCR transgenic | The Jackson Laboratory | Cat# 003831 |
| ||
Oligonucleotides | ||
| ||
See Table S1 for list of quantitative RT-PCR primers | This paper | N/A |
| ||
Recombinant DNA | ||
| ||
Plasmid: MIG-EV | Xiong et al., 202051 | N/A |
Plasmid: MIG-Ahr | Xiong et al., 202051 | N/A |
Plasmid: MIG-Y9A | Xiong et al., 202051 | N/A |
Plasmid: MIG-ΔbHLH | Xiong et al., 202051 | N/A |
Plasmid: hCD2-EV | Laboratory of Dr. Weishan Huang | N/A |
Plasmid: hCD2-Blimp1 | Laboratory of Dr. Weishan Huang | N/A |
| ||
Software and algorithms | ||
| ||
FlowJo version 10.4.2 | FlowJo | https://www.flowjo.com |
Prism 8 | GraphPad Software | https://www.graphpad.com/scientific-software/prism/ |
Highlights.
Ahr acts as a promoter of resident memory CD8+ T cell differentiation and function
Ahr suppresses the circulating but promotes the resident memory core gene program
Ahr enhances polyfunctional CD8+ T cells, which drive anti-tumor immunity
In human, AHR promotes in vitro TRM differentiation and granzyme B production
ACKNOWLEDGMENTS
We thank the entire Zhou and Avram labs for their help and suggestions. We thank John Bostick for analytical assistance as well as Shainal Gandhi and Lauren Dee for their technical assistance. We thank the Flow Cytometry and ICBR core facilities at the University of Florida and the Genomics Facility at the University of Chicago for sequencing service and assistance. Funding: This work was supported by the NIH (R01AI132391 and R01AI157109, L.Z.; R01AI067846 and 5P30CA076292, D.A.; J.W.D was partially supported by T32AI007110). L.Z. was a Pew Scholar in Biomedical Sciences, supported by the Pew Charitable Trusts, and is an Investigator in the Pathogenesis of Infectious Disease, supported by Burroughs Wellcome Fund. This work was made possible, in part, by NIH Instrumentation Grant 1S10 OD021676–01.
Footnotes
DECLARATION OF INTERESTS
The authors declare they have no competing interests.
SUPPLEMENTAL INFORMATION
Supplemental information can be found online at https://doi.org/10.1016/j.celrep.2022.111963.
REFERENCES
- 1.Liu L, Gong T, Tao W, Lin B, Li C, Zheng X, Zhu S, Jiang W, and Zhou R (2019). Commensal viruses maintain intestinal intraepithelial lymphocytes via noncanonical RIG-I signaling. Nat. Immunol. 20, 1681–1691. [DOI] [PubMed] [Google Scholar]
- 2.Szabo PA, Miron M, and Farber DL (2019). Location, location, location: tissue resident memory T cells in mice and humans. Sci. Immunol 4, eaas9673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Sheridan BS, Pham QM, Lee YT, Cauley LS, Puddington L, and Lefranç ois L (2014). Oral infection drives a distinct population of intestinal resident memory CD8(+) T cells with enhanced protective function. Immunity 40, 747–757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Williams MA, and Bevan MJ (2007). Effector and memory CTL differentiation. Annu. Rev. Immunol. 25, 171–192. [DOI] [PubMed] [Google Scholar]
- 5.Jameson SC, and Masopust D (2018). Understanding subset diversity in T cell memory. Immunity 48, 214–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kaech SM, and Cui W (2012). Transcriptional control of effector and memory CD8+ T cell differentiation. Nat. Rev. Immunol. 12, 749–761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Arsenio J, Metz PJ, and Chang JT (2015). Asymmetric cell division in T lymphocyte fate diversification. Trends Immunol. 36, 670–683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Flossdorf M, Rössler J, Buchholz VR, Busch DH, and Höfer T (2015). CD8+ T cell diversification by asymmetric cell division. Nat. Immunol. 16, 891–893. [DOI] [PubMed] [Google Scholar]
- 9.Youngblood B, Hale JS, Kissick HT, Ahn E, Xu X, Wieland A, Araki K, West EE, Ghoneim HE, Fan Y, et al. (2017). Effector CD8 T cells dedifferentiate into long-lived memory cells. Nature 552, 404–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Bannard O, Kraman M, and Fearon DT (2009). Secondary replicative function of CD8+ T cells that had developed an effector phenotype. Science 323, 505–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Yuzefpolskiy Y, Baumann FM, Kalia V, and Sarkar S (2015). Early CD8 T-cell memory precursors and terminal effectors exhibit equipotent in vivo degranulation. Cell. Mol. Immunol. 12, 400–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Joshi NS, Cui W, Chandele A, Lee HK, Urso DR, Hagman J, Gapin L, and Kaech SM (2007). Inflammation directs memory precursor and short-lived effector CD8(+) T cell fates via the graded expression of T-bet transcription factor. Immunity 27, 281–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.MacKay LK, Rahimpour A, Ma JZ, Collins N, Stock AT, Hafon ML, Vega-Ramos J, Lauzurica P, Mueller SN, Stefanovic T, et al. (2013). The developmental pathway for CD103+ CD8+ tissue-resident memory T cells of skin. Nat. Immunol. 14, 1294–1301. [DOI] [PubMed] [Google Scholar]
- 14.Obar JJ, and Lefranç ois L (2010). Early signals during CD8 T cell priming regulate the generation of central memory cells. J. Immunol. 185, 263–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Kaech SM, Tan JT, Wherry EJ, Konieczny BT, Surh CD, and Ahmed R (2003). Selective expression of the interleukin 7 receptor identifies effector CD8 T cells that give rise to long-lived memory cells. Nat. Immunol. 4, 1191–1198. [DOI] [PubMed] [Google Scholar]
- 16.Herndler-Brandstetter D, Ishigame H, Shinnakasu R, Plajer V, Stecher C, Zhao J, Lietzenmayer M, Kroehling L, Takumi A, Kometani K, et al. (2018). KLRG1+ effector CD8+ T cells lose KLRG1, differentiate into all memory T cell lineages, and convey enhanced protective immunity. Immunity 48, 716–729.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Gerlach C, Moseman EA, Loughhead SM, Alvarez D, Zwijnenburg AJ, Waanders L, Garg R, de la Torre JC, and von Andrian UH (2016). The chemokine receptor CX3CR1 defines three antigen-experienced CD8 T cell subsets with distinct roles in immune surveillance and homeostasis. Immunity 45, 1270–1284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Milner JJ, Nguyen H, Omilusik K, Reina-Campos M, Tsai M, Toma C, Delpoux A, Boland BS, Hedrick SM, Chang JT, and Goldrath AW (2020). Delineation of a molecularly distinct terminally differentiated memory CD8 T cell population. Proc. Natl. Acad. Sci. USA 117, 25667–25678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Mueller SN, Gebhardt T, Carbone FR, and Heath WR (2013). Memory T cell subsets, migration patterns, and tissue residence. Annu. Rev. Immunol. 31, 137–161. [DOI] [PubMed] [Google Scholar]
- 20.Sallusto F, Lenig D, Förster R, Lipp M, and Lanzavecchia A (1999). Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 401, 708–712. [DOI] [PubMed] [Google Scholar]
- 21.Sathaliyawala T, Kubota M, Yudanin N, Turner D, Camp P, Thome JJC, Bickham KL, Lerner H, Goldstein M, Sykes M, et al. (2013). Distribution and compartmentalization of human circulating and tissue-resident memory T cell subsets. Immunity 38, 187–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Steinert EM, Schenkel JM, Fraser KA, Beura LK, Manlove LS, Igyártó BZ, Southern PJ, and Masopust D (2015). Quantifying memory CD8 T cells reveals regionalization of immunosurveillance. Cell 161, 737–749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Schenkel JM, Fraser KA, Vezys V, and Masopust D (2013). Sensing and alarm function of resident memory CD8+ T cells. Nat. Immunol. 14, 509–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ariotti S, Hogenbirk MA, Dijkgraaf FE, Visser LL, Hoekstra ME, Song JY, Jacobs H, Haanen JB, and Schumacher TN (2014). T cell memory. Skin-resident memory CD8⁺ T cells trigger a state of tissue-wide pathogen alert. Science 346, 101–105. [DOI] [PubMed] [Google Scholar]
- 25.Schenkel JM, Fraser KA, Beura LK, Pauken KE, Vezys V, and Masopust D (2014). T cell memory. Resident memory CD8 T cells trigger protective innate and adaptive immune responses. Science 346, 98–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kok L, Masopust D, and Schumacher TN (2021). The precursors of CD8+ tissue resident memory T cells: from lymphoid organs to infected tissues. Nat. Rev. Immunol. 22, 283–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Milner JJ, and Goldrath AW (2018). Transcriptional programming of tissue-resident memory CD8 + T cells. Curr. Opin. Immunol. 51, 162–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Milner JJ, Toma C, Yu B, Zhang K, Omilusik K, Phan AT, Wang D, Getzler AJ, Nguyen T, Crotty S, et al. (2017). Runx3 programs CD8+ T cell residency in non-lymphoid tissues and tumours. Nature 552, 253–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Mackay LK, Minnich M, Kragten NAM, Liao Y, Nota B, Seillet C, Zaid A, Man K, Preston S, Freestone D, et al. (2016). Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes. Science 352, 459–463. [DOI] [PubMed] [Google Scholar]
- 30.Rutishauser RL, Martins GA, Kalachikov S, Chandele A, Parish IA, Meffre E, Jacob J, Calame K, and Kaech SM (2009). Transcriptional repressor Blimp-1 promotes CD8+ T cell terminal differentiation and represses the acquisition of central memory T cell properties. Immunity 31, 296–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Fonseca R, Beura LK, Quarnstrom CF, Ghoneim HE, Fan Y, Zebley CC, Scott MC, Fares-Frederickson NJ, Wijeyesinghe S, Thompson EA, et al. (2020). Developmental plasticity allows outside-in immune responses by resident memory T cells. Nat. Immunol. 21, 412–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Behr FM, Parga-Vidal L, Kragten NAM, van Dam TJP, Wesselink TH, Sheridan BS, Arens R, van Lier RAW, Stark R, and van Gisbergen KPJM (2020). Tissue-resident memory CD8+ T cells shape local and systemic secondary T cell responses. Nat. Immunol. 21, 1070–1081. [DOI] [PubMed] [Google Scholar]
- 33.Cervantes-Barragan L, Chai JN, Tianero MD, Di Luccia B, Ahern PP, Merriman J, Cortez VS, Caparon MG, Donia MS, Gilfillan S, et al. (2017). Lactobacillus reuteri induces gut intraepithelial CD4+CD8αα+ T cells. Science 357, 806–810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zelante T, Iannitti RG, Cunha C, De Luca A, Giovannini G, Pieraccini G, Zecchi R, D’Angelo C, Massi-Benedetti C, Fallarino F, et al. (2013). Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 39, 372–385. [DOI] [PubMed] [Google Scholar]
- 35.Gutiérrez-Vázquez C, and Quintana FJ (2018). Regulation of the immune response by the aryl hydrocarbon receptor. Immunity 48, 19–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ye J, Qiu J, Bostick JW, Ueda A, Schjerven H, Li S, Jobin C, Chen ZME, and Zhou L (2017). The aryl hydrocarbon receptor preferentially marks and promotes gut regulatory T cells. Cell Rep. 21, 2277–2290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Maekawa Y, Minato Y, Ishifune C, Kurihara T, Kitamura A, Kojima H, Yagita H, Sakata-Yanagimoto M, Saito T, Taniuchi I, et al. (2008). Notch2 integrates signaling by the transcription factors RBP-J and CREB1 to promote T cell cytotoxicity. Nat. Immunol. 9, 1140–1147. [DOI] [PubMed] [Google Scholar]
- 38.Burrows K, Antignano F, Bramhall M, Chenery A, Scheer S, Korinek V, Underhill TM, and Zaph C (2017). The transcriptional repressor HIC1 regulates intestinal immune homeostasis. Mucosal Immunol. 10, 1518–1528. [DOI] [PubMed] [Google Scholar]
- 39.Crowl JT, Heeg M, Ferry A, Milner JJ, Omilusik KD, Toma C, He Z, Chang JT, and Goldrath AW (2022). Tissue-resident memory CD8+ T cells possess unique transcriptional, epigenetic and functional adaptations to different tissue environments. Nat. Immunol. 23, 1121–1131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Bai A, Hu H, Yeung M, and Chen J (2007). Krüppel-like factor 2 controls T cell trafficking by activating L-selectin (CD62L) and sphingosine-1-phosphate receptor 1 transcription. J. Immunol. 178, 7632–7639. [DOI] [PubMed] [Google Scholar]
- 41.Mackay LK, Wynne-Jones E, Freestone D, Pellicci DG, Mielke LA, Newman DM, Braun A, Masson F, Kallies A, Belz GT, and Carbone FR (2015). T-Box transcription factors combine with the cytokines TGF-β and IL-15 to control tissue-resident memory T cell fate. Immunity 43, 1101–1111. [DOI] [PubMed] [Google Scholar]
- 42.Skon CN, Lee JY, Anderson KG, Masopust D, Hogquist KA, and Jameson SC (2013). Transcriptional downregulation of S1pr1 is required for the establishment of resident memory CD8+ T cells. Nat. Immunol. 14, 1285–1293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Casey KA, Fraser KA, Schenkel JM, Moran A, Abt MC, Beura LK, Lucas PJ, Artis D, Wherry EJ, Hogquist K, et al. (2012). Antigen-independent differentiation and maintenance of effector-like resident memory T cells in tissues. J. Immunol. 188, 4866–4875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Boivin WA, Cooper DM, Hiebert PR, and Granville DJ (2009). Intracellular versus extracellular granzyme B in immunity and disease: challenging the dogma. Lab. Invest. 89, 1195–1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Solovyev VV, Shahmuradov IA, and Salamov AA (2010). Identification of promoter regions and regulatory sites. Methods Mol. Biol. 674, 57–83. [DOI] [PubMed] [Google Scholar]
- 46.Wang S, Sun H, Ma J, Zang C, Wang C, Wang J, Tang Q, Meyer CA, Zhang Y, and Liu XS (2013). Target analysis by integration of transcriptome and ChIP-seq data with BETA. Nat. Protoc. 8, 2502–2515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Li S, Bostick JW, Ye J, Qiu J, Zhang B, Urban JF Jr., Avram D, and Zhou L (2018). Aryl hydrocarbon receptor signaling cell intrinsically inhibits intestinal group 2 innate lymphoid cell function. Immunity 49, 915–928.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Minsavage GD, Park SK, and Gasiewicz TA (2004). The aryl hydrocarbon receptor (AhR) tyrosine 9, a residue that is essential for AhR DNA binding activity, is not a phosphoresidue but augments AhR phosphorylation. J. Biol. Chem. 279, 20582–20593. [DOI] [PubMed] [Google Scholar]
- 49.Swanson HI, and Yang JH (1996). Mapping the protein/DNA contact sites of the Ah receptor and Ah receptor nuclear translocator. J. Biol. Chem. 271, 31657–31665. [DOI] [PubMed] [Google Scholar]
- 50.Evans BR, Karchner SI, Allan LL, Pollenz RS, Tanguay RL, Jenny MJ, Sherr DH, and Hahn ME (2008). Repression of Aryl Hydrocarbon Receptor (AHR) signaling by AHR repressor: role of DNA binding and competition for AHR nuclear translocator. Mol. Pharmacol. 73, 387–398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Xiong L, Dean JW, Fu Z, Oliff KN, Bostick JW, Ye J, Chen ZE, Mühlbauer M, and Zhou L (2020). Ahr-Foxp3-RORgt axis controls gut homing of CD4+ T cells by regulating GPR15. Sci. Immunol. 5, eaaz7277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Gálvez-Cancino F, López E, Menares E, Díaz X, Flores C, Cáceres P, Hidalgo S, Chovar O, Alcántara-Hernández M, Borgna V, et al. (2018). Vaccination-induced skin-resident memory CD8+ T cells mediate strong protection against cutaneous melanoma. Oncoimmunology 7, e1442163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Mami-Chouaib F, Blanc C, Corgnac S, Hans S, Malenica I, Granier C, Tihy I, and Tartour E (2018). Resident memory T cells, critical components in tumor immunology. J. Immunother. Cancer 6, 87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Murray IA, Patterson AD, and Perdew GH (2014). Aryl hydrocarbon receptor ligands in cancer: friend and foe. Nat. Rev. Cancer 14, 801–814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Xue P, Fu J, and Zhou Y (2018). The aryl hydrocarbon receptor and tumor immunity. Front. Immunol. 9, 286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Szabo PA, Levitin HM, Miron M, Snyder ME, Senda T, Yuan J, Cheng YL, Bush EC, Dogra P, Thapa P, et al. (2019). Single-cell transcriptomics of human T cells reveals tissue and activation signatures in health and disease. Nat. Commun. 10, 4706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Zaid A, Mackay LK, Rahimpour A, Braun A, Veldhoen M, Carbone FR, Manton JH, Heath WR, and Mueller SN (2014). Persistence of skin-resident memory T cells within an epidermal niche. Proc. Natl. Acad. Sci. USA 111, 5307–5312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Lawrence BP, Roberts AD, Neumiller JJ, Cundiff JA, and Woodland DL (2006). Aryl hydrocarbon receptor activation impairs the priming but not the recall of influenza virus-specific CD8+ T cells in the lung. J. Immunol. 177, 5819–5828. [DOI] [PubMed] [Google Scholar]
- 59.Winans B, Nagari A, Chae M, Post CM, Ko CI, Puga A, Kraus WL, and Lawrence BP (2015). Linking the aryl hydrocarbon receptor with altered DNA methylation patterns and developmentally induced aberrant antiviral CD8+ T cell responses. J. Immunol. 194, 4446–4457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Goudot C, Coillard A, Villani AC, Gueguen P, Cros A, Sarkizova S, Tang-Huau TL, Bohec M, Baulande S, Hacohen N, et al. (2017). Aryl hydrocarbon receptor controls monocyte differentiation into dendritic cells versus macrophages. Immunity 47, 582–596.e6. [DOI] [PubMed] [Google Scholar]
- 61.Liu Y, Zhou N, Zhou L, Wang J, Zhou Y, Zhang T, Fang Y, Deng J, Gao Y, Liang X, et al. (2021). IL-2 regulates tumor-reactive CD8+ T cell exhaustion by activating the aryl hydrocarbon receptor. Nat. Immunol. 22, 358–369. [DOI] [PubMed] [Google Scholar]
- 62.Liu Y, Liang X, Dong W, Fang Y, Lv J, Zhang T, Fiskesund R, Xie J, Liu J, Yin X, et al. (2018). Tumor-repopulating cells induce PD-1 expression in CD8+ T cells by transferring kynurenine and AhR activation. Cancer Cell 33, 480–494.e7. [DOI] [PubMed] [Google Scholar]
- 63.Prasad S, Hu S, Sheng WS, Chauhan P, Singh A, and Lokensgard JR (2017). The PD-1: PD-L1 pathway promotes development of brain-resident memory T cells following acute viral encephalitis. J. Neuroinflammation 14, 82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Li C, Zhu B, Son YM, Wang Z, Jiang L, Xiang M, Ye Z, Beckermann KE, Wu Y, Jenkins JW, et al. (2019). The transcription factor Bhlhe40 programs mitochondrial regulation of resident CD8+ T cell fitness and functionality. Immunity 51, 491–507.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Liikanen I, Lauhan C, Quon S, Omilusik K, Phan AT, Bartrolí LB, Ferry A, Goulding J, Chen J, Scott-Browne JP, et al. (2021). Hypoxia-inducible factor activity promotes antitumor effector function and tissue residency by CD8+ T cells. J. Clin. Invest. 131, e143729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Boyd A, Almeida JR, Darrah PA, Sauce D, Seder RA, Appay V, Gorochov G, and Larsen M (2015). Pathogen-specific T cell polyfunctionality is a correlate of T cell efficacy and immune protection. PLoS One 10, e0128714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.De Groot R, Van Loenen MM, Guislain A, Nicolet BP, Freen-Van Heeren JJ, Verhagen OJHM, Van Den Heuvel MM, De Jong J, Burger P, Van Der Schoot CE, et al. (2019). Polyfunctional tumor-reactive T cells are effectively expanded from non-small cell lung cancers, and correlate with an immune-engaged T cell profile. Oncoimmunology 8, e1648170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Egelston CA, Avalos C, Tu TY, Simons DL, Jimenez G, Jung JY, Melstrom L, Margolin K, Yim JH, Kruper L, et al. (2018). Human breast tumor-infiltrating CD8+ T cells retain polyfunctionality despite PD-1 expression. Nat. Commun. 9, 4297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Bowcutt R, Malter LB, Chen LA, Wolff MJ, Robertson I, Rifkin DB, Poles M, Cho I, and Loke P (2015). Isolation and cytokine analysis of lamina propria lymphocytes from mucosal biopsies of the human colon. J. Immunol. Methods 421, 27–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Fernandez-Salguero P, Pineau T, Hilbert DM, McPhail T, Lee SS, Kimura S, Nebert DW, Rudikoff S, Ward JM, and Gonzalez FJ (1995). Immune system impairment and hepatic fibrosis in mice lacking the dioxin-binding Ah receptor. Science 268, 722–726. [DOI] [PubMed] [Google Scholar]
- 71.Qiu J, Heller JJ, Guo X, Chen Z.m.E., Fish K, Fu YX, and Zhou L (2012). The aryl hydrocarbon receptor regulates gut immunity through modulation of innate lymphoid cells. Immunity 36, 92–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Fu Z, Dean JW, Xiong L, Dougherty MW, Oliff KN, Chen ZME, Jobin C, Garrett TJ, and Zhou L (2021). Mitochondrial transcription factor A in RORgt+ lymphocytes regulate small intestine homeostasis and metabolism. Nat. Commun. 12, 4462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, and Gingeras TR (2013). STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Li B, and Dewey CN (2011). RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics 12, 323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Love MI, Huber W, and Anders S (2014). Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, and Mesirov JP (2005). Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, Puigserver P, Carlsson E, Ridderstråle M, Laurila E, et al. (2003). PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273. [DOI] [PubMed] [Google Scholar]
- 78.R Core Team (R Foundation for Statistical Computing, Vienna, Austria. 2022). R: A Language and Environment for Statistical Computing. [Google Scholar]
- 79.Murdoch D, and Adle D rgl: 3D Visualization Using OpenGL. R package. 2021. [Google Scholar]
- 80.Kolde R (R package. 2019). Pheatmap: Pretty Heatmaps. [Google Scholar]
- 81.Langmead B, and Salzberg SL (2012). Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, and Durbin R; 1000 Genome Project Data Processing Subgroup (2009). The sequence alignment/map format and SAMtools. Bioinformatics 25, 2078–2079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Quinlan AR, and Hall IM (2010). BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Heinz S, Benner C, Spann N, Bertolino E, Lin YC, Laslo P, Cheng JX, Murre C, Singh H, and Glass CK (2010). Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Butler A, Hoffman P, Smibert P, Papalexi E, and Satija R (2018). Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Trapnell C, Cacchiarelli D, Grimsby J, Pokharel P, Li S, Morse M, Lennon NJ, Livak KJ, Mikkelsen TS, and Rinn JL (2014). The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
All data supporting the findings of this study are available within the article and its supplementary information and from the corresponding author upon reasonable request. The accession number of the RNA-Seq, ChIP-Seq, and scRNA-Seq data reported in this paper have been uploaded to GEO at the accession number: GSE220944. This paper does not report original code. Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.