Abstract Celiac disease (CD) is an autoimmune disease in which intestinal inflammation is induced by dietary gluten. The means through which gluten-specific CD4^+ T cell activation culminates in intraepithelial T cell (T-IEL) mediated intestinal damage remain unclear. Here, we performed multiplexed-single cell analysis of intestinal and gluten-induced peripheral blood T cells from patients in different celiac disease states and healthy controls. Untreated, active CD (ACD) and potential CD (PCD) were associated with an enrichment of activated intestinal T cell populations including CD4^+ follicular T-helper (T[FH]) cells, regulatory T cells (Tregs), and natural CD8^+ αβ and γδ T-IELs. Natural CD8^+ αβ and γδ T-IELs expressing activating natural killer cell receptors (NKRs) exhibited a distinct TCR repertoire in CD, which persisted in patients on a gluten-free diet (GFD) without intestinal inflammation. Our data further show that NKR-expressing cytotoxic cells, which appear to mediate intestinal damage in CD, arise from a distinct NKR-expressing memory population of T-IELs. Following gluten ingestion, both αβ and γδ T cell clones from this memory population of T-IELs circulated systemically along with gluten-specific CD4^+ T cells and assumed a cytotoxic and activating NKR-expressing phenotype. Collectively, these findings suggest that cytotoxic T cells in CD are rapidly mobilized in parallel with gluten-specific CD4^+ T cells following gluten ingestion. One Sentence Summary: Gluten ingestion induces cytotoxic T-IELs from an NKR-expressing natural memory population in parallel with gluten-specific CD4^+ T cells. INTRODUCTION Celiac disease (CD) is a common autoimmune disease characterized by dietary gluten-induced small intestinal mucosal injury in HLA-DQ2 or HLA-DQ8 individuals ([64]1). Beyond a lifelong gluten-free diet (GFD), current treatment options for CD are limited. In addition to HLA association, CD shares many hallmarks with other autoimmune diseases, including non-MHC genetics, immune-mediated tissue destruction, and auto-antibody production ([65]2). However, CD is one of the only autoimmune diseases for which the triggering antigen is well-defined and can be controlled, and thus provides an opportunity to study autoimmune T cell responses in different disease states. How gluten-dependent CD4^+ T cell activation culminates in intestinal epithelial cell (IEC) destruction remains incompletely understood ([66]3–[67]5). Gluten-specific CD4^+ T cell immunity is necessary for disease but is insufficient for IEC destruction in humans or in mouse models ([68]3, [69]6, [70]7). Instead, intestinal intraepithelial T cells (T-IEL) are thought to be the end effector cells that drive tissue damage in CD ([71]5). In patients with potential CD (PCD), characterized by positive serum celiac antibodies in the absence of mucosal damage, CD4^+ T cell responses are intact but appear to be uncoupled from T-IEL-mediated tissue damage ([72]6, [73]7). A substantial portion of PCD patients eventually develop CD, while others remain disease-free for years despite continued gluten consumption ([74]7). An increase in T-IELs is a pathologic hallmark of CD, and these cells persist in celiac mucosa despite GFD ([75]8). T-IELs in CD have not been demonstrated to recognize gluten and their cytolytic activity can be induced irrespective of antigen specificity ([76]8). They also express activating natural killer cell receptors (NKRs), which are considered critical for IEC destruction ([77]4, [78]9–[79]12). These findings have led to the prevailing model, which proposes that T-IELs within tissues are activated by stress and inflammation, presumably initiated by gluten-specific CD4^+ T cells ([80]4, [81]8). Re-introducing dietary gluten to celiac patients on a GFD rapidly induces gluten-specific CD4^+ T cells in the peripheral blood that express activation markers as well as the intestinal-trafficking β7 integrin receptor ([82]13). This phenomenon likely represents the induction of a memory immune response to gluten, as seen in the peripheral blood en route from gut-associated lymphoid tissues to the intestine. In addition to gluten-specific CD4^+ T cells, oral gluten challenge leads to a parallel induction of activated, gut-homing CD8^+ αβ and γδ T cells in the peripheral blood ([83]14). These cells phenotypically resemble IELs, express CD103, and share a TCR repertoire with intestinal T-IELs ([84]14, [85]15). However, the significance of these findings is unclear as the functional significance of gluten-induced circulating CD8^+ αβ and γδ T cells has not been established. Here, we leveraged the specific opportunities afforded to us with individuals who are in different states of CD and can be re-exposed to gluten to dissect mechanisms of autoimmunity using single-cell multiomics. We analyzed T cell responses and TCR repertoire at the site of T cell-driven autoimmunity in the presence or absence of the gluten trigger. Through oral gluten challenge, we were able to study the induction of CD through tracking intestinal T cell clones in the peripheral blood. RESULTS Single-cell profiling of duodenal T cells in CD We obtained duodenal biopsies from 11 patients with newly diagnosed or active CD (ACD), 19 celiac patients treated with a gluten-free diet (GFD), 7 PCD patients, and 17 healthy controls ([86]Fig. 1A). Marsh scores, which range from 0 (normal) to 3 (most severe), are used to grade disease severity in celiac biopsies through assessment of villous height and immune infiltration. All ACD patients had modified Marsh 3a-3c scores, indicative of IEL infiltration and villous atrophy, while GFD/PCD patients had Marsh 0 (17/19 GFD, 6/7 PCD patients) or Marsh 1 (2/19 GFD, 1/7 PCD patients) scores ([87]table S1) ([88]16). PCD patients were HLA-DQ2/8 with positive celiac serology (anti-tissue-transglutaminase 2 (TG2) IgA) in the absence of intestinal damage by histology ([89]table S1) ([90]6, [91]7). Live CD3^+ cells were flow sorted, and scRNA-seq, scTCR-seq, and 204 panel CITE-seq (cellular indexing of transcriptomes and epitopes by sequencing) were performed using the droplet-based 10X Genomics platform ([92]Fig. 1A, [93]fig. S1A, [94]tables S1 and [95]S2). Following quality control preprocessing to remove unidentifiable cells, multiplets, and low-quality cells, 41,285 intestinal T cells remained for analysis. Fig 1. Distinct CD4^+ Treg and T[FH] cell phenotypes enriched in untreated CD mucosa. Fig 1. [96]Open in a new tab (A) Schematic representation of experimental design and sorting strategy. (B-D) UMAP plot of intestinal T cells colored by (B) patient condition, (C) annotated cell type, and (D) identified clusters with T[FH] and Treg subclusters highlighted (encircled). (E) Quantification of CD4^+ T cells within T[FH] and Treg subclusters in control (n = 17), PCD (n = 7), ACD (n = 11), and GFD (n = 19) mucosa. (F) Heatmap of mean RNA (left) and ADT (right; ADT-stained samples only) expression of selected genes and cell surface markers identified through differential expression across T[FH] and Treg subclusters and remaining CD4^+ T cells. (G) Heatmap of mean RNA expression of select genes, identified through differential expression, within T[FH] subcluster cells across states of CD. (H) Scatterplot comparing percentage of T[FH] and Treg subcluster cells in untreated CD (n = 18) mucosa. Shaded region indicates 95% confidence interval following linear regression. Two-sided Wilcoxon rank sum test (E). *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.001. Pearsons rank correlation coefficient (H). To resolve CD3^+ T cell subtypes, we used scTCR-seq, CITE-seq antibody derived tags (ADTs), and reference-based scRNA-seq cell type recognition to annotate CD4^+ TCRαβ (CD4^+), CD8^+ TCRαβ (CD8^+), and γδ T cells corroborating relative percentages determined during FACS sorting ([97]fig. S1, [98]A to [99]C) ([100]17–[101]19). Transcriptomic data from all patient samples was integrated using canonical correlation analysis, and dimensionality reduction was performed to visualize unsupervised clustering of single cells as Uniform Manifold Approximation and Projection (UMAP) embeddings ([102]Fig. 1, [103]B and [104]C, [105]fig. S1, [106]D to [107]F). Eight clusters were identified and annotated by means of distinct transcriptomic signatures and cell surface marker expression ([108]Fig. 1D, [109]fig. S1G, [110]table S3). CD4^+ T cell populations were identified as T-helper 1 (Th1), T-helper 17 (Th17), and a shared cluster of follicular T-helper (T[FH]) and regulatory T cells (Tregs) (T[FH]/Treg cluster) ([111]Fig. 1, [112]C and [113]D). CD8^+ T cell populations were classified as effector memory (T[EM]), resident memory IELs (T[RM]), and natural IELs (natural-memory; T[NM], natural-effector; T[NE]) ([114]20), which shared phenotype with γδ T cells ([115]Fig. 1, [116]C and [117]D). We also observed a small cluster of naïve T cells, containing both CD4^+ and CD8^+ T cells ([118]Fig. 1, [119]C and [120]D). Induction of activated CD4^+ T cell populations in CD The study of CD4^+ T cells in CD has largely focused on gluten-specific T cells, which were reported to have a distinctive T[FH] phenotype ([121]21). Several groups have also reported an increase in FOXP3^+ T cells in active celiac mucosa ([122]22, [123]23). We found that a distinct population of activated CD4^+ T cells (T[FH]/Treg cluster), comprised of both T[FH]-phenotype and FOXP3^+CD25^+CD4^+ Tregs, was highly enriched within CD mucosa relative to controls ([124]Fig. 1, [125]B to [126]D, [127]fig. S2A, [128]table S4). The T[FH]/Treg cluster contained activated T cells with elevated expression of CD39, TIGIT, CCR4, and ICOS and low expression of CD49a, IL7R, and CD73 ([129]fig. S2B). We subclustered the T[FH]/Treg cluster, which yielded a subcluster of Tregs and a subcluster of T[FH]-phenotype T cells ([130]Fig. 1D). Both activated T[FH] and Treg-phenotype cells were enriched in ACD and PCD relative to GFD and controls ([131]Fig. 1E). T[FH]-subcluster CD4^+ T cells matched the previously reported, pro-inflammatory phenotype of gluten tetramer-reactive cells ([132]21), expressing CXCL13, CD200, CD84, IFNG, IL-21, PD-1, CD39, and β7-integrin ([133]Fig. 1F). We also observed an upregulation of transcription factors (TFs) BCL6 and LEF1, both shown to be critical for T[FH] differentiation ([134]Fig. 1F) ([135]24, [136]25). Notably, T[FH]-subcluster CD4^+ T cells in inflamed ACD mucosa had elevated expression of IFNG relative to T[FH]-subcluster cells in non-inflamed PCD mucosa, which highly expressed IL-21 ([137]Fig. 1G). While FOXP3-expressing CD4^+ T cells have now been well-described in CD, their functional significance is unclear ([138]26, [139]27). Consistent with a regulatory role, cells within the Treg subcluster exhibited an activated and suppressive phenotype, expressing CD39, GITR, and CD27 in addition to IL10, CD25, CTLA4, and PD-L1 ([140]Fig. 1F) ([141]28, [142]29). This CD39^+TIGIT^+IL10^+ phenotype matches that of Tregs recently described in Crohn’s disease ([143]30). To investigate the regulatory networks characterizing the distinct T[FH]and Treg phenotypes, we performed gene co-expression analysis followed by the identification of TF cis-regulatory motifs ([144]31, [145]32). 310 active TF modules were found and scored individually for each cell. T[FH] and Treg subclusters shared several active TF modules including LEF-1, BATF, ETV7 and c-MAF ([146]fig. S2C). In contrast, other CD4^+ T cell populations used RORγt, RORα, KLF3, and RUNX2 TF networks ([147]fig. S2C). It has been reported that circulating gluten-induced CD4^+ T cells in CD co-expressed FOXP3 and CD39, suggesting that both effector and regulatory CD4^+ T cell responses in CD may be gluten-specific ([148]26). We investigated the TCR repertoire across T[FH] and Treg subclusters. In CD mucosa, T[FH]-phenotype cells exhibited elevated usage of V-gene biases previously identified in gluten-specific CD4^+ T cells ([149]fig. S2D) ([150]33–[151]36). We next applied the GLIPH2 algorithm (grouping of lymphocyte interaction by paratope hotspots) to cluster TCRs by potential epitope specificity ([152]37). Within the T[FH] cluster, GAET, LRGG, R(X)PSTDT, and QNTG motifs were identified and shared across multiple patients ([153]table S5). Within the LRGG motif, we observed TCRs utilizing arginine at CDR3β position 5, previously implicated in binding the immunodominant gliadin α-II epitope ([154]35, [155]38) ([156]table S5). Collectively, these results corroborate the presence of gluten-specific CD4^+ T cells within the T[FH] population. Within the Treg subcluster, different CDR3β motifs, including S(X)YRE, GALS, and SLGLAGA(X)E, were enriched, and there was negligible sharing between T[FH] and Treg subclusters, suggesting they are selected by different antigens and arguing against the gluten specificity of Tregs ([157]table S5). Despite distinct TCR repertoires, we observed a strong correlation between the percentage of CD4^+ T cells exhibiting T[FH] and Treg phenotypes within untreated CD patients, suggesting both populations are induced together in CD ([158]Fig. 1H). Accordingly, in PCD, where gluten-induced CD4^+ T cell responses are intact, both T[FH] and Treg populations were enriched ([159]Fig. 1, [160]E and [161]H). Natural CD8^+ αβ and γδ T-IELs exhibit an NK-polarized phenotype We next examined populations of intestinal CD8^+ and γδ T cells ([162]Fig. 1, [163]C and [164]D). We observed the presence of effector memory CD8^+ T cells (T[EM] cluster; CD28^+CD44^+) in addition to several populations of CD103^+ T-IELs, which we termed resident memory (T[RM], CD73^+IL7R^+), natural-memory (T[NM,] CD38^+CD39^+NKR^+IL7R^+) and natural-effector (T[NE,] CD38^+CD39^+NKR^+IL7R^−) ([165]Fig. 1D, [166]Fig. 2, [167]A to [168]C, [169]fig. S1G). Notably, T[NM] and T[NE] populations contained both CD8^+ and “naturally-occurring” γδ T-IELs ([170]Fig. 1C) ([171]20). Fig 2. Natural αβ and γδ T-IELs exhibit NK-reprogramming in CD. Fig 2. [172]Open in a new tab (A) Dotplot heatmap showing mean expression of select ADT markers across identified clusters (ADT-stained samples only). (B) ADT expression profile of selected cluster-defining cell surface markers (ADT-stained samples only). (C) RNA expression profile of selected cluster-defining marker genes. (D) UMAP plot of intestinal T cells colored by AUC scores of individual cells for selected canonical pathway activities. (E) Quantification of CD8^+ and γδ T cells across identified T[RM], T[EM], and natural clusters in control (n = 17), PCD (n = 7), ACD (n = 11), and GFD (n = 19) mucosa. Two-sided Wilcoxon rank sum test (E). *P < 0.05, **P < 0.01, and ***P < 0.001. T[RM] cluster T-IELs expressed CD73, CD127, and CD103, in combination with high expression of KLRC1 (NKG2A) ([173]Fig. 2, [174]A to [175]C). Consistent with the capacity of NKG2A to restrict T cell activation ([176]39), T[RM] T-IELs scored highly for gene ontology (GO) pathways involved in negatively regulating both T cell immunity and cytotoxicity ([177]Fig. 2D). These cells comprised the largest fraction of CD8^+ T cells from healthy controls, and as such, represent the “baseline” CD8^+ T-IEL population within healthy duodenum ([178]Fig. 2E). T[EM] cluster cells exhibited a highly cytotoxic phenotype, expressing GZMK, EOMES, NKG2D, PD-1, and KLRG1 with limited CD103 expression, suggestive of their presence within the lamina propria (LP) as opposed to the epithelium ([179]Fig. 2, [180]A to [181]C, [182]fig. S3A) ([183]20). Cytotoxic CD8^+ T cells expressing EOMES and KLRG1 have been implicated in mediating tissue damage in inflammatory bowel disease (IBD) ([184]32, [185]40). However, we did not observe an enrichment of T[EM] cells across CD disease states suggesting they do not similarly drive tissue damage in CD ([186]Fig. 2E, [187]fig. S3B). T-IELs that we termed “natural” highly expressed activating NKRs, including KLRC2 (NKG2C), KLRC3 (NKG2E), and KLRC4 (NKG2F), in addition to killer immunoglobulin-like receptors (KIRs), including KIR2DL4, analogous to previously described “natural” murine T-IELs ([188]20, [189]39) ([190]Fig. 2C, [191]fig. S3, [192]C and [193]D). Natural T-IEL populations exhibited GO signatures consistent with NK polarization ([194]3, [195]4, [196]9–[197]12), including NK cell-mediated immunity, cytokine production, and NKR-driven interactions with non-classical MHC class I molecules ([198]39) ([199]Fig. 2D, [200]fig. S3A). Notably, we observed an increased frequency of “natural” CD8^+ T-IELs in all celiac disease states, including PCD ([201]Fig. 2E). Natural T-IEL populations define stages of CD Given the importance of NKR-expressing T-IEL populations in CD ([202]3, [203]4, [204]9–[205]12), we re-clustered 28,380 CD8^+ and γδ T-IELs, and identified ten distinct clusters of CD8^+ and γδ T-IELs ([206]Fig. 3, [207]A and [208]B, [209]fig. S4, [210]A to [211]C). Subsequent analysis was performed on T-IELs, which were distinguished through expression of CD103 ([212]20). Fig 3. Distinct Natural T-IEL phenotypes define CD progression. Fig 3. [213]Open in a new tab (A-B) UMAP plot of intestinal CD8^+ and γδ T-IELs colored by (A) patient condition and (B) identified clusters. (C) Heatmap of mean RNA expression of selected genes identified through differential expression across natural T-IEL clusters and remaining CD8^+ and γδ T-IELs. (D) Quantification of CD8^+ and γδ T-IELs across natural T-IEL clusters in control (n = 17), PCD (n = 7), ACD (n = 11), and GFD (n = 19) mucosa. (E) Heatmap of mean RNA expression of select genes, identified through differential expression, within T[NE] T-IELs across states of CD. (F) Volcano plot of differentially expressed genes (log2FC expression) between γδ (left) and CD8^+ (right) T[NE] T-IELs from (n = 37) CD patients. (G) Scatterplot comparing percentage of CD8^+ and γδ T-IELs in CD (n = 37) mucosa across select natural clusters. Shaded region indicates 95% confidence interval following linear regression. Two-sided Wilcoxon rank sum test (D). *P < 0.05, **P < 0.01, and ***P < 0.001. Pearsons rank correlation coefficient (G). We observed substantial changes within NKR-expressing populations across different stages of CD. Within the previously described natural-memory population, we observed three distinct clusters, which we termed Innate-like (Innate), natural-memory 1 (T[NM]1), and natural-memory 2 (T[NM]2) ([214]Fig. 3B). The most abundant population of γδ T-IELs in healthy mucosa (T[NM]1 cluster) expressed TGFB1 (TGF-β), AREG (Amphiregulin), the TFs ID2 and ID3, and 4–1BB, suggestive of immuno-regulatory and tissue repair-promoting roles ([215]Fig. 3, [216]B to [217]D, [218]fig. S4D) ([219]41). Therefore, as T[RM] (described above) represents the baseline CD8^+ T cell phenotype ([220]Fig. 2, [221]A to [222]E, [223]fig. S4C), T[NM]1 similarly represents the baseline γδ T cell phenotype in healthy duodenum. Despite being the baseline γδ T cell phenotype, there were clearly CD8^+ T cells present within the T[NM]1 cluster ([224]Fig. 3D, [225]fig. S4A). In contrast to γδ T cells, there were relatively few T[NM]1-phenotype CD8^+ T cells in healthy control mucosa compared to celiac mucosa ([226]Fig. 3D). However, both T[NM]1-phenotype CD8^+ and γδ T cells were less abundant within ACD mucosa compared to non-inflamed PCD and GFD mucosa ([227]Fig. 3D). This population, which has not been well characterized previously, represents a distinct memory T-IEL population, as the TF ID3 is critical for long-lived memory CD8^+ T cell generation ([228]42). In healthy mucosa, we also observed the presence of γδ T-IELs (Innate cluster) highly expressing TYROBP (DAP12), CD247, and SH2D1B in addition to elevated expression of CD122 and natural cytotoxicity receptors (NCRs) NKp46 and NKp44 ([229]Fig. 3, [230]A to [231]D, [232]fig. S4D). These innate-like γδ T-IELs were diminished in CD mucosa and highly utilized TRGV4, matching a previously described population of Vδ1^+ γδ T-IELs lost in CD mucosa with disease onset ([233]Fig. 3D, [234]fig. S4E) ([235]43). It was previously reported that a distinct Vδ1^+ γδ T-IEL population persists in celiac patients despite GFD, consistent with “permanent reshaping” or “scarring” of T-IELs in CD ([236]43). Consistent with this report, T-IELs exhibiting a similar NKR-expressing phenotype (T[NM]2 cluster), were significantly increased in both ACD and GFD, irrespective of gluten exposure or intestinal damage ([237]Fig. 3, [238]C and [239]D). In addition to Vδ1^+ γδ T-IELs, we clearly observed both CD8^+ and non-Vδ1^+ (~52% of T[NM]2 γδ T cells) T-IELs within this population ([240]Fig. 3D, [241]fig. S4, [242]A and [243]F). Both αβ and γδ T-IELs within this T[NM]2 population exhibited similar enrichment in CD mucosa relative to controls ([244]Fig. 3D), and highly expressed activating NKRs, KLRC2, KLRC3, and KLRC4, and markers indicative of cytotoxic potential including HCST (DAP10) and KLRK1 (NKG2D) ([245]Fig. 3, [246]C and [247]D). However, despite their cytotoxic potential, this population was present and enriched within GFD mucosa which lacked any discernable tissue damage, suggesting that they were not actively mediating cytotoxicity ([248]Fig. 3D). Within inflamed ACD mucosa, we observed a population of pro-inflammatory, IFNG-expressing CD8^+ and γδ T-IELs, which we termed natural-effector (T[NE]) ([249]Fig 3, [250]A to [251]D). T[NE] T-IELs exhibited a highly cytotoxic and activated phenotype through expression of activating NKR KLRC2 in addition to PRF1, NKG7, GZMB, CD71, CD52, and CD161 ([252]Fig. 3C, [253]fig. S4D). Their highly cytotoxic phenotype and enrichment in ACD mucosa strongly suggest that T[NE] T-IELs are responsible for IEC destruction in CD ([254]Fig. 3, [255]A to [256]D). Interestingly, T[NE] T-IELs were also enriched in PCD relative to GFD or control mucosa, in the absence of histologic tissue damage ([257]Fig. 3D). Compared to ACD, T[NE] T-IELs in PCD were present at lower frequency and exhibited reduced expression of PRF1, IFNG, KLRC2, KLRC3, and KLRC4 relative to ACD T[NE] T-IELs ([258]Fig. 3, [259]D and [260]E). These findings imply additional mechanisms of T[NE] activation may differentiate ACD from PCD ([261]3–[262]5, [263]11, [264]44). Aside from their TCR genes, CD8^+ and γδ T-IELs within natural T-IEL populations were phenotypically indistinguishable, suggestive of redundant roles ([265]Fig. 3F, [266]fig. S4G, [267]table S6). Within T[NE], T[NM]1, and T[NM]2 T-IEL clusters, we observed a moderate to strong correlation between the percentage of CD8^+ and γδ T-IELs exhibiting each phenotype, implying these NKR-expressing CD8^+ αβ and γδ populations are induced together in CD ([268]Fig. 3G). Distinct natural T-IEL TCR αβ and γδ repertoires in CD We next analyzed scTCR-seq data to investigate both the CD8^+ αβ and γδ TCR repertoires across different CD states. Across identified populations, we observed substantial variations in the degree of clonal expansion in both CD8^+ and γδ T cells ([269]Fig. 4, [270]A and [271]B, [272]fig. S5, [273]A to [274]D). Interestingly, in healthy mucosa, “baseline” populations (CD8^+, T[RM] 1–4; γδ, T[NM]1), contained elevated fractions of highly expanded TCR clones (33.1% αβ, 46.4% γδ) ([275]Fig. 4, [276]A and [277]B). Conversely, natural (T[NM]1, T[NM]2, and T[NE]) T-IELs, the predominant αβ and γδ T-IEL populations in CD mucosa, were clonally diverse in CD, affirming previously reported findings of increased clonal diversity in CD ([278]Fig. 4, [279]A and [280]B) ([281]45, [282]46). Fig 4. Distinct TCR repertoire of Natural αβ and γδ T-IELs in CD. Fig 4. [283]Open in a new tab (A-B) (A) CDR3β and (B) CDR3δ clonal expansion quartiles across and select clusters within healthy (HC) and CD mucosa. (C-D) UpSet plot displaying shared (C) αβ or (D) γδ clonotypes across T-IEL clusters within untreated CD mucosa. Barplot on left indicates count of unique (C) CDR3β or (D) CDR3δ within clusters. Shared clonotype pairing is visualized as a black circle joined by black lines to indicate sharing sets, with number of unique shared (C) CDR3β or (D) CDR3δ between clusters (independent of expansion) annotated above the barplot. Highlighted bars indicate sharing between natural clusters. (E-F) Chord diagram displaying TRGV-TRDV pairing usage across natural T-IEL clusters within (E) control and (F) CD mucosa. We examined TCR sharing across clusters within untreated CD mucosa, where we observed ~15% of unique clonotypes were present across multiple clusters (570/3,737 shared/total αβ clonotypes; 151/971 shared/total γδ clonotypes) ([284]Fig. 4, [285]C and [286]D). Broadly, within untreated CD mucosa containing all natural, T[RM], and T[EM] populations, natural T-IELs showed extensive TCR repertoire sharing with other natural populations, as opposed to T[RM] and T[EM] populations ([287]Fig. 4, [288]C and [289]D). For both CD8^+ and γδ T-IELs, we observed frequent TCR sharing between T[NM]1 and T[NM]2 clusters (Most frequent αβ, 140 clonotypes, Most frequent γδ, 81 clonotypes) and T[NM]2 and T[NE] populations (2^nd most frequent αβ, 50 clonotypes; 2^nd most frequent γδ, 18 clonotypes) ([290]Fig. 4, [291]C and [292]D). TCRγδ sharing between innate and natural (T[NM]1, T[NM]2, and T[NE]) populations was very limited, suggesting these populations arise independently ([293]Fig. 4D). Thus, our TCR data show that in CD, natural T-IEL populations, including T[NM]1, T[NM]2 and T[NE], extensively shared TCR repertoire with one another, but not with T[RM], T[EM], or Innate populations. Accordingly, we frequently observed clones present within each of the T[NM]1, T[NM]2 and T[NE] clusters for both αβ (27 clonotypes) and γδ (11 clonotypes) T-IELs ([294]Fig. 4, [295]C and [296]D). Among γδ T-IELs, healthy controls preferentially utilized TRDV3 (64.5% Control; 32.2% CD) compared to CD patients, who preferentially utilized TRDV1 (25.1% Control; 50.9% CD) ([297]fig. S5, [298]E to [299]G). In both healthy and CD mucosa, innate T-IELs exhibited a semi-invariant usage of both TRGV4/TRDV1 and TRGV4/TRDV3, and were found at levels consistent with those of previously described butyrophilin-reactive Vδ1^+ γδ T-IELs ([300]Fig. 3D, [301]Fig. 4, [302]E and [303]F) ([304]43). In contrast, within natural clusters, CD patients exhibited highly distinct TRGV/TRDV pairings when compared to healthy controls, with increased diversity of pairings observed in CD mucosa ([305]Fig. 4, [306]E and [307]F). As such, natural T-IELs in CD exhibited greater αβ and γδ clonal diversity when compared to healthy controls ([308]Fig. 4, [309]A and [310]B). Collectively, these results suggest TCR-mediated selection of disease-associated γδ T-IELs in CD. CITE-seq clustering of intestinal T cells To further profile cell surface marker repertoire of previously described populations, protein level data from a 204 antibody CITE-seq panel across ADT-stained patient samples (n = 26,066 IEL and LP T cells) was integrated using reciprocal principal component analysis to cluster intestinal T cells by cell surface marker phenotype ([311]fig. S6, [312]A and [313]B and [314]table S1). CITE-seq largely recaptured scRNAseq-based populations of intestinal T cells ([315]fig. S6C). We again observed that activated T[FH] and Treg populations (CITE-seq Tfh/Treg cluster) were highly enriched in ACD and clustered distinctly from other CD4^+ T cell populations ([316]fig. S6, [317]B to [318]D). Confirming our transcriptomic analysis, healthy mucosa was enriched with KLRC1^+CD73^+CD103^+CD8^+ T[RM] cells, while cytotoxic KLRG1-expressing T[EM] cells did not show significant changes across disease states ([319]fig. S6, [320]A to [321]G). Increased levels of activated CD8^+ T cells expressing CD38, CD39, CD71, IFNG, and PRF1 (CITE-seq T[NE] cluster) were again observed in ACD mucosa ([322]fig. S6, [323]A to [324]G). While surface protein level clustering was unable to fully resolve scRNA-seq-defined T[NM]1 and T[NM]2 T-IEL clusters, we observed an enrichment of “natural” CD38^+CD39^+NKR^+CD8^+ T-IELs (CITE-seq T[NM] cluster) in GFD mucosa, providing further evidence for the persistence of natural, NKR-expressing CD8^+ T cells in CD mucosa ([325]fig. S6, [326]B to [327]H). Gluten challenge induces rapid reprogramming of natural CD8^+ and γδ T-IELs Following oral gluten challenge in CD patients on a GFD, CD8^+ and γδ T cells expressing the activation marker CD38 and gut-homing integrins CD103 and β7 integrin appear in peripheral blood ([328]14). Their functional significance, however, remains unclear ([329]47). We analyzed 3,924 CD38^+CD103^+ CD8^+ and γδ T cells from five patients at day 6 following three-day oral gluten challenge ([330]Fig. 5A, [331]fig. S7, [332]A and [333]B). Four of the five patients underwent intestinal biopsy prior to gluten challenge, enabling us to track the phenotype of shared T cell clones in the intestine and peripheral blood ([334]Fig. 5A, [335]fig. S7C, [336]table S1). The relative frequency of gluten-induced CD38^+CD103^+CD8^+ (0.16–1.3%) and CD38^+CD103^+γδ (0.08–3.73%) T cells in the peripheral blood varied across patients, consistent with prior reports ([337]fig. S7B) ([338]14). Fig 5. Gluten challenge induces circulating T[NE]-phenotype cells from intestinal T[NM]1 clones. Fig 5. [339]Open in a new tab (A) Schematic representation of experimental design and sorting strategy of peripheral blood, gluten-induced (CD38^+CD103^+) CD8^+ and γδ T cells. (B-C) Projection of transcriptomic profile of gluten-induced T cells onto UMAP plot of intestinal T-IELs colored by (B) gluten challenge sample and (C) predicted cluster using unimodal UMAP projection. (D) Quantification of predicted cluster of gluten-induced circulating T cells from (n = 5) gluten challenge patients. (E) RNA expression profile of selected gluten-induced T cell-enriched genes identified through differential expression within gluten-induced circulating and intestinal CD8^+ and γδ T cells. (F) UMAP plot colored by shared TCR clones of pre-challenge intestinal and post-challenge peripheral blood T cells, split by predicted phenotype of gluten-induced T cells. (G) Volcano plot of differentially expressed genes (log2FC expression) between shared TCR clones of pre-challenge intestinal (left) and post challenge peripheral blood (right) T[NE] T-IELs from (n = 4) CD patients. (H) Chord diagram displaying TRGV-TRDV pairing usage within gluten-induced γδ T cells. Two-sided Wilcoxon rank sum test (D), **P < 0.01. To investigate the phenotype of these gluten-induced peripheral blood T cells, we projected their transcriptomic profile atop our previously described intestinal CD8^+ and γδ T-IEL populations using unimodal UMAP projection ([340]Fig. 5B). Gluten-induced CD38^+CD103^+ CD8^+ and γδ T cells exhibited a highly cytotoxic phenotype consistent with T[NE] intestinal T cells, highly enriched in untreated CD ([341]Fig. 5, [342]B to [343]E, [344]fig. S7D). These cells similarly exhibited a pathogenic, NK-reprogrammed phenotype, highly upregulating IFNG, PRF1, CD52, KLRC2, HCST, KLRK1, in addition to MKI67 (KI67) and cell surface expression of CD94 ([345]Fig. 5E, [346]fig. S7, [347]E and [348]F). Confirming a prior report, gluten-induced CD8^+ and γδ T cells also highly expressed activation markers observed among T[FH] cells including HLA-DR and CD71 ([349]Fig. 1F, [350]fig. S7F) ([351]47). Thus, gluten induces circulating CD8^+ and γδ T cells in peripheral blood exhibiting a NK-reprogrammed cytotoxic T[NE] phenotype, similar to T-IELs enriched in PCD and ACD mucosa. For patients who underwent both biopsy and gluten challenge, we observed significant sharing of αβ and γδ TCRs across the intestine and peripheral blood ([352]15). Interestingly, we found that these common clones had distinct phenotypes in intestine vs. peripheral blood, suggestive of gluten-induced reprogramming ([353]Fig. 5, [354]F and [355]G, [356]table S7). Intestinal αβ and γδ T-IEL clones from the T[NM]1 cluster, the baseline γδ T cell population in healthy controls, were later seen in the blood exhibiting the cytotoxic T[NE] phenotype ([357]Fig. 3, [358]B to [359]D, [360]Fig. 5F, [361]table S8). While we also observed a limited number of intestinal T[EM] and T[RM] clones circulating in peripheral blood after gluten challenge, in contrast to natural T-IELs, these clones largely retained their intestinal phenotype in the blood ([362]Fig. 5F, [363]table S8). Therefore, as suggested by TCR sharing between T[NM] and T[NE] T-IELs within untreated CD mucosa ([364]Fig. 4, [365]C and [366]D), these results from gluten challenge affirm the common ancestry of natural-memory and natural-effector αβ and γδ populations. These data further show that intestinal αβ and γδ T cell clones present in the T[NM]1 cluster undergo rapid reprogramming upon gluten ingestion, assuming the cytotoxic and pro-inflammatory T[NE] phenotype in the peripheral blood. It was previously reported that gluten-induced circulating CD8^+ and γδ T cell populations show TCR similarity and clones are recalled upon repeat gluten challenge, suggesting antigen-driven selection ([367]14). To assess the possibility that these gluten-induced peripheral blood T cells were selected by specific antigens, we used the GLIPH2 algorithm to cluster TCRs to identify motifs critical for antigen-binding ([368]37). We observed convergence within the CDR3β region of gluten-induced CD8^+ T cells and identified numerous motifs shared across patients ([369]table S9). Further, like natural celiac γδ T-IELs, gluten-induced circulating γδ T cells did not show the biased TRGV usage seen in natural T-IELs from healthy mucosa ([370]Fig. 4, [371]E and [372]F, [373]Fig. 5H, and [374]fig. S5G). Collectively these results suggest that natural αβ and γδ T-IELs in CD are selected by antigens distinct from natural T-IELs in healthy mucosa. Although gluten recognition by T-IELs has not been demonstrated, these circulating αβ and γδ T[NE] T-IELs are induced by gluten with the same kinetics as gluten-specific CD4^+ T cells ([375]8, [376]14). DISCUSSION Here, we leveraged opportunities afforded in CD to comprehensively study T cell phenotype and TCR repertoire across different disease states in CD. As CD shares traits with other autoimmune diseases, these findings should prove informative beyond CD. Intestinal gluten-reactive CD4^+ T cells were previously ascribed a T[FH]-phenotype through mass cytometry and bulk RNAseq ([377]21). Through our analysis, we identified a similar population of CD4^+ T cells, which were enriched in untreated CD mucosa, but largely absent in controls. Consistent with gluten-reactivity, we observed motifs seen in gluten-reactive TCRs and biased variable gene usage within this population ([378]33–[379]36, [380]38). The T[FH] phenotype of these cells are consistent with the idea that gluten-specific T cells interact with antigen-specific B cells (anti-gliadin, anti-TG2) serving as antigen presenting cells, reciprocally amplifying humoral and cell-mediated immune responses in CD ([381]48). These T[FH] cells phenotypically resembled those reported in rheumatoid arthritis and Crohn’s disease, and thus may have a similar role in these diseases ([382]30, [383]49). Tregs have been shown to be expanded within CD mucosa ([384]22, [385]23), but their functional significance is unclear ([386]27). Our data show CD39^+CD25^+FOXP3^+ Tregs in CD shared activation markers and TF networks with T[FH]-phenotype cells, but exhibited a suppressive phenotype (IL10, CD25, CTLA4, PD-L1). We further observed a positive correlation between the relative frequencies of T[FH] cells and Tregs in ACD and PCD mucosa, suggesting they are induced together. It was reported that gluten challenge induces circulating gluten-specific CD39^+FOXP3^+ Tregs ([387]26), although a more recent study demonstrated that FOXP3-expressing gluten-specific T cells do not co-express CD25 or phenotypically resemble Tregs ([388]21). Our TCR data show established gluten-reactive motifs present in T[FH] but not Treg cells, and negligible repertoire overlap between the two populations. Therefore, our data are consistent with the idea that inflammatory (T[FH]) and regulatory CD4^+ T cells are induced together in CD through gluten exposure, but are selected by different antigens. The importance of activating NKR-expressing T-IELs in celiac pathogenesis has now been well established ([389]4, [390]8–[391]12). The current prevailing model proposes that these T-IELs, which are responsible for tissue damage, are activated downstream of CD4^+ T cell-driven inflammation ([392]5, [393]8). Our data provide several novel insights regarding how NKR-expressing T-IEL responses are modulated in CD. An increase in T-IELs is a pathologic hallmark of CD, and known to persist even with dietary gluten exclusion. We describe two populations of NKR-expressing, natural memory T-IEL, T[NM]1 and T[NM]2, that are modulated in CD. While celiac-associated T-IEL populations were previously reported to consist of Vδ1^+ γδ T cells ([394]43), we found both CD8^+ αβ and non-Vδ1^+ γδ T-IELs present across all CD-modulated natural populations. The T[NM]1 population, the most abundant population of γδ T-IELs in healthy mucosa, expressed TGFB1 and AREG suggestive of immuno-regulatory and tissue repair-promoting roles. The T[NM]2 phenotype exhibited an NK-reprogrammed phenotype and highly upregulated HCST and KLRK1, suggestive of cytotoxic potential. Despite their cytotoxic potential, T[NM]2 T-IELs were relatively enriched in non-inflamed CD mucosa, suggesting they were not actively mediating intestinal damage. Our analysis instead implicates cells within the natural-effector (T[NE]) cluster as the cells responsible for tissue damage in CD. This cluster contained activated T-IELs with a highly cytotoxic and pro-inflammatory phenotype, expressing PRF1, GZMB, and IFNG, and was significantly enriched in damaged ACD mucosa. While this population was predominantly comprised of CD8^+ T-IELs, we also observed the presence of phenotypically indistinguishable γδ T[NE] T-IELs, suggesting redundant roles of natural CD8^+ and γδ T-IELs in CD. These natural-effector cells extensively shared TCR repertoire with CD-enriched natural-memory T-IELs, consistent with the prevailing model whereby exposure to IL-15 and CD4^+ T cell-mediated inflammation induces activation of NKR-expressing, natural-memory T-IELs to assume the fully cytotoxic natural-effector phenotype ([395]2–[396]5, [397]8, [398]44). Notably, we also observed an enrichment of natural-effector T-IELs in noninflamed PCD mucosa when compared to both control and GFD patients. When compared to ACD mucosa, T[NE] T-IELs were present at lower levels in PCD mucosa and expressed lower levels of pro-inflammatory mediators. While it is possible that a threshold level of T[NE] T-IELs is necessary for tissue damage and not reached in PCD, we speculate that mucosal expression of IL-15 and markers of epithelial stress, which distinguish PCD from ACD, may be required to unleash the inflammatory potential of T[NE] T-IELs ([399]3–[400]5, [401]11, [402]44). Our TCR data show that in CD, natural-memory and natural-effector cells show evidence of TCR-mediated selection and shared TCR repertoire, establishing that these populations share ancestry. One caveat to our study is that we were unable to obtain biopsies from the same patients in different disease states, limiting our ability to infer directionality of differentiation of T cell populations that share TCR repertoire. However, we were able to obtain gluten-challenged peripheral blood from four patients that previously underwent intestinal biopsy. These data establish directionality and timing of differentiation, showing that intestinal natural-memory clones, predominantly from the T[NM]1 phenotype, are reprogrammed to assume the natural-effector phenotype rapidly, within 6 days, upon gluten ingestion. This reprogramming occurs in parallel with gluten-specific CD4^+ T cell activation, before significant intestinal inflammation is seen histologically, suggesting it occurs deliberately and not downstream of CD4^+ T cell-induced inflammation ([403]14). Further evidence of the gluten-induced transition from T[NM]1 to natural-effector phenotype is seen in the diminished frequency of T[NM]1 cells in ACD compared to GFD mucosa. Beyond showing TCR repertoire sharing, our data do not show directionality of differentiation between T[NM]2 and T[NM]1/T[NE] populations. We speculate that once reprogrammed T[NE] cells are recruited to the intestine through gluten exposure, clones are retained as T[NM]2 T-IELs, which can be re-induced to mediate cytotoxicity upon gluten re-encounter. Although T-IELs in CD are not thought to recognize gluten themselves ([404]8), their activation may still be dependent on gluten-specific CD4^+ T cells. Multiple aspects of effector CD8^+ T cell responses are known to depend upon CD4^+ T cell help ([405]50–[406]52). We propose that gluten-specific CD4^+ T cells similarly license the activation and recruitment of cytotoxic natural-effector T-IELs in CD. This licensing may occur via IL-21, implicated in CD8^+ T cell migration and effector function ([407]53), which was highly produced by gluten-reactive CD4^+ T cells ([408]8, [409]21). Interestingly, IL-21R was highly expressed in both T[NM]1 T-IELs and Tregs in CD. If gluten-specific CD4^+ T cells do indeed interact with T[NM]1 CD8^+ T cells, this interaction would likely occur in gut-associated lymphoid tissue, as these two cell types reside in different locations within the intestine (epithelium vs. lamina propria). As our study was entirely performed on dissociated tissues, spatial context is lacking and we relied on established markers to infer the presence of cells in the intestinal epithelium ([410]20). The question of how gluten induces parallel inflammatory responses by gluten-specific CD4^+ T cells and T-IELs that have not been demonstrated to recognize gluten is an important one, and likely applicable in other HLA class II-associated autoimmune diseases ([411]2). A similar phenomenon of parallel activation of circulating CD4^+, CD8^+ and γδ T cells was reported in a murine model of CD4^+ T cell-initiated autoimmunity ([412]54). In this model, circulating CD8^+ T cells functioned as regulatory CD8^+ T cells, suppressing proliferation of autoreactive CD4^+ T cells ([413]54). In contrast, our data strongly suggest that gluten-induced circulating natural CD8^+ and γδ T cells in CD have a pro-inflammatory, rather than regulatory, role. We note, however, that the pro-inflammatory role we ascribe to natural CD8^+ and γδ T cells in CD and the regulatory role seen in the mouse model are not mutually exclusive. One possibility that reconciles these two models is that natural CD8^+ and γδ T-IEL populations are self-reactive. Indeed, T-IEL populations in humans and mice have been shown to display self-reactive properties ([414]20, [415]55, [416]56). Their self-reactivity would enable both rapid mobilization to kill tissue in response to infection as well as feedback inhibition of pro-inflammatory CD4^+ T cells. MATERIALS AND METHODS Study Design The goal of our study was to leverage unique traits of celiac disease to study the recruitment, activation, and phenotypic alterations of T cells across distinct states of disease, at the single-cell level. To this end, we generated a single-cell multiomics dataset from T cells isolated from duodenal biopsies of eleven ACD, nineteen GFD, seven PCD, and seventeen control patients in addition to gluten-induced peripheral blood T cells from five CD patients following oral gluten challenge (see [417]table S1). Human Specimens Duodenal biopsies from patients undergoing endoscopy at Columbia University Irving Medical Center were collected with informed consent, as approved by the Columbia University IRB (AAAB2472). All celiac disease patients had a biopsy-confirmed diagnosis. Patients were categorized as potential celiac disease based on positive TTG serology and HLA-DQ2/8 in the absence of histologic findings of celiac disease ([418]7). Gluten-free diet patients were treated for at least 6 months and had confirmed mucosal healing based on pathology. Peripheral blood from volunteers who underwent oral gluten challenge was collected with informed consent, as approved by the Columbia University IRB (AAAR1608). For gluten challenge, volunteers with biopsy-proven CD, adherent to a strict GFD for at least 6 months, consumed four slices of white bread (~15g gluten) per day for 3 consecutive days (days 1–3), and donated blood on day 6, as described ([419]14). Biopsy Sample Processing Fresh tissue was dissociated into single-cell suspensions using sequential enzymatic digestions to isolate both IELs (1x HBSS, 1mM EDTA, 1mM DTT, and 5% FBS: 30 min shaking) and LPLs (1x HBSS, 5% FBS, and 0.025mg/mL Liberase: 90 min shaking) or with Multi Tissue Dissociation Kit 1 (Miltenyi Biotec) ([420]Table S1). Single-cell suspensions were cryopreserved (40% FBS 10% DMSO) and stored in liquid nitrogen. Peripheral Blood Mononuclear Cell Isolation Whole blood was collected in BD Vacutainer Collection tubes. Peripheral blood mononuclear cells were isolated by density gradient centrifugation (Ficoll-Paque, Fisher Scientific). Single-cell suspensions were cryopreserved (40% FBS 10% DMSO) and stored in liquid nitrogen. CITE-Seq Antibody-Oligo Hyperconjugation Conjugation of purified antibodies for cell hashing was performed as described ([421]https://cite-seq.com/protocol/). Antibody clones and barcodes are available in [422]table S2. CITE-seq Cryopreserved single-cell suspensions were thawed at 37°C, aliquots of IEL/LPL fraction pooled (when applicable), and rested for 8 hours in RPMI/10% FBS. Single-cell suspensions were incubated with Fc-receptor block for 10 minutes at 4°C (TruStain FcX, BioLegend) followed by hashing antibody (0.5μg per 2 million cells) for 30 minutes at 4°C. Following multiple washes, samples were stained with anti-CD3 (clone OKT3, BioLegend) FITC, anti-CD4 (clone SK3, BD-Horizon) BUV737, anti-CD8α (clone RPA-T8, BioLegend) BV785, anti-CD45 (clone HI30, BioLegend) BV711, and anti-TCRγδ (clone 5A6.E9, Thermo Fisher) PE. Gluten challenge samples were stained with anti-CD38 (clone HIT2, BioLegend) APC, anti-CD103 (clone Ber-ACT8, BioLegend) BV711, and anti-Integrin β7 (clone FIB504, BioLegend) PE/Dazzle^™ 594. Samples were incubated with pre-titrated TotalSeq-C CITE-seq antibody cocktail (BioLegend, PN900000115) for 30 minutes at 4°C. LIVE/DEAD Fixable Red (ThermoFisher) and DAPI were used to gate viable cells. T cells were sorted into RPMI by FACS AriaII or Propel Labs Bigfoot Cell Sorter by gating viable CD3^+ intestinal cells and CD3^+CD4^−CD38^+CD103^+ cells from gluten challenge samples. FACS analysis performed using FlowJo (v.10.7.1) software. Droplet-based CITE-seq FACS-sorted T cells were loaded onto the Chromium 10x Genomics V1 or v1.1 platform (10X Genomics) per manufacturer’s instructions. Single cell 5’ transcript, TCRαβ, and CITE-seq libraries were prepared using Chromium Single Cell 5’ V(D)J with Feature Barcoding Reagent Kits ([423]CG000186) or Chromium Next GEM Single Cell V(D)J Reagent Kits v1.1 with Feature Barcode Technology for Cell Surface Protein ([424]CG000208). Cell hashtag library generation was performed as described ([425]https://cite-seq.com/protocol/). TCRγδ libraries were prepared as described ([426]17). cDNA libraries were quantified using High Sensitivity D5000 ScreenTape (Agilent) and sequenced using Illumina MiSeq, NextSeq 550, and NovaSeq 6000 platforms. Data Processing To process gene expression and CITE-seq sequencing reads, Cell Ranger v5.0.0 software (10x Genomics) was used. Raw base call files were demultiplexed using cellranger mkfastq and aligned to GRCh38 reference genome and CITE-seq barcode list for 5’ mRNA and CITE-seq respectively with cellranger count. TCRαβ, and TCRγδ sequencing reads were aligned to GRCh38 reference genome with cellranger vdj version 3.0.2 (10x Genomics). Raw cell hashtag reads were processed using CITE-seq-Count to generate cell hashing count matrix ([427]https://github.com/Hoohm/CITE-seq-Count). Hashes were normalized to total counts across cells (total UMI counts set at 100). Cell barcode identity was assigned to most highly expressed hashtag that encompassed a minimum of 2/3 the recovered UMIs. Shared CDR3 nucleotide sequence was used to rescue unidentified cells. Remaining unidentified cells were removed from subsequent analysis. Multimodal Single-cell Analysis Preprocessing and clustering analysis steps were performed using the Seurat R package. The percentage of genes mapping to mitochondria, ribosomes, and total UMIs were used to remove “low-quality” cells (>15% mitochondrial genes OR < 350 genes detected). Global-scaling normalization and identification of 2000 most variable genes were performed prior to integration across runs using canonical correlation analysis available in the Seurat R package. A second round of filtering was performed to remove additional “low-quality” (mitochondrial-gene-expressing, ribosomal-gene-expressing, HSP-gene-expressing, cycling cells) clusters of cells. CITE-seq count matrix was normalized using centered log-ratio transformation following adjustment to UMI counts to account for isotype controls in an isotype-specific manner. CITE-seq clustered data was integrated using reciprocal PCA analysis as available in the Seurat R package. Cells were visualized as UMAP embeddings and clustered with resolutions of 0.2 for all intestinal T cells, 0.55 for CD8^+ and γδ T-IELs, and 0.35 for CITE-seq-clustered data. Subclustering was performed on UMAP derived all intestinal T cells on a shared cluster T[FH]/Treg/T-Naïve cluster to isolate a cluster of naïve T cells. Subclustering was performed on T-IEL UMAP T[RM] cluster to isolate additional subpopulations of T[RM] T-IELs. Gluten-induced peripheral blood T-IELs were additionally filtered using CITE-seq ADTs (GC 1–3) and scRNA-seq (GC 1–5) to remove cells that did not express CD38, CD103, and β7-Integrin. Unimodal UMAP projection was performed to project transcriptomic profile of gluten-induced T-IELs onto UMAP structure of intestinal CD8^+ and γδ T-IELs as available in the Seurat R package. For TCR sharing, shared T cell clones were assigned phenotype (T[NE]/T[NM]1/T[NM]2/T[EM]/T[RM]/MAIT/Innate) based on most abundant predicted cell phenotypes per clone. Identification of Cell Types TCRαβ and TCRγδ cells were annotated based on scTCR-seq output. Remaining T cells without a detected TCR were annotated as αβ or γδ T cells through CITE-seq expression of TCRαβ and TCRγδ in addition to expression of TCRγδ genes (i.e. TRDC, TRGC1, TRDV1). αβ T cells were then annotated as CD4^+ or CD8^+ based on combination of CITE-seq CD4 and CD8 antibodies followed by annotation with the SingleR package with Monaco Immune Dataset for unbiased reference-based scRNA-seq cell type annotation. T-IELs were annotated through transcriptomic and CITE-seq expression of CD103. scRNA-seq Differential Expression Analysis Differential expression between populations was performed using the R package MAST via the Seurat wrapper function FindMarkers. Minimum log-fold threshold was set to default parameters (0.25) with exception of generation of volcano plots where minimum log-fold threshold was set to 0. Differentially expressed genes available in [428]Supplementary Materials. TCR Analysis TCRαβ and TCRγδ sequences annotated as “productive” were kept for further analysis. TCR counts were based on CDR3β, CDR3δ, CDR3γ amino acid sequences in a patient-dependent manner. Expanded TCRs were split into quartiles based on clonal frequency. Variable gene pairings visualized using the chordDiagram function available in the circlize R package. Sharing of CDR3β and CDR3δ sequences across clusters visualized using the UpSetR R package. CDR3β, TRBV gene, TRBJ gene, and CDR3α sequence were input into the GLIPH V2 algorithm ([429]http://50.255.35.37:8080/) using cell type matched reference. Fisher-exact test performed to assess confidence of clusters. Gene Ontology and Pathway Enrichment Analysis Individual cells were scored for pathway activities using area-under-the-curve (AUC) analysis with the AUCell R package. Gene expression rankings for each cell was created using the AUCell_buildRankings function with default parameters. The canonical pathway database was downloaded from the Broad Institute, with gene sets used to score each cell individually for each gene set using calculated AUC values with the function AUCell_calcAUC. Gene Regulatory Network Analysis The R package SCENIC was used to examine transcription factor module activity across intestinal T cells. Log-transformed count matrix was filtered to exclude genes detected in fewer than 50 cells, minimum counts per gene less than 130 (~1 average UMI count across all cells), and genes not detected in RcisTarget databases. Positive and negative target associations between transcription factors and genes were identified using the runCorrelation function on the filtered count matrix. Gene regulatory network analysis using stochastic Gradient Boosting Machine regression with early-stopping regularization was performed using GRNBoost2, as available in the arboreto python program. Detected coexpression modules for transcription factors were scored using AUCell package as previously described in “[430]Gene Ontology and Pathway Enrichment Analysis”. Statistical Analysis Statistical tests performed in [431]Fig. 1E, [432]1H, [433]2E, [434]3D, [435]3G, [436]5D, [437]figs. S2A, [438]S3B, [439]S4B, and [440]S6D, were performed in R using the ggpubr package. Differential expression was performed in R using Seurat and MAST packages. Statistical tests performed in [441]tables S5 and [442]S9 used GLIPH2 program. Unpaired Mann-Whitney tests were performed for non-parametric data (min. 10 CD4^+, CD8^+, γδ T cells/patient). Pearson rank correlation coefficient in [443]Figs. 1H and [444]3G. [445]Table S4 contains data input into statistical calculations. Fisher’s exact test was used to determine enrichment of GLIPH2-identified TCR motifs. Supplementary Material Table S3 [446]NIHMS1924047-supplement-Table_S3.xlsx^ (10.1KB, xlsx) Table S2 [447]NIHMS1924047-supplement-Table_S2.xlsx^ (21.6KB, xlsx) Table S4 [448]NIHMS1924047-supplement-Table_S4.xlsx^ (81.1KB, xlsx) Table S5 [449]NIHMS1924047-supplement-Table_S5.xlsx^ (17.7KB, xlsx) main supplementary [450]NIHMS1924047-supplement-main_supplementary.pdf^ (1.7MB, pdf) Table S8 [451]NIHMS1924047-supplement-Table_S8.xlsx^ (9.8KB, xlsx) Table S9 [452]NIHMS1924047-supplement-Table_S9.xlsx^ (47.4KB, xlsx) Table S6 [453]NIHMS1924047-supplement-Table_S6.xlsx^ (1.5MB, xlsx) Table S7 [454]NIHMS1924047-supplement-Table_S7.xlsx^ (611.8KB, xlsx) MDAR checklist [455]NIHMS1924047-supplement-MDAR_checklist.docx^ (68.2KB, docx) Table S1 [456]NIHMS1924047-supplement-Table_S1.xlsx^ (14.1KB, xlsx) Acknowledgements: