The NOD mouse develops spontaneous type 1 diabetes, with some features of disease that are very similar to the human disease. However, a proportion of NOD mice are naturally protected from developing diabetes, and currently, studies characterizing this cohort are very limited. Here, using both immunofluorescence and multiparameter flow cytometry, we focus on the pancreatic islet morphology and immune infiltrate observed in naturally protected NOD mice. We show that naturally protected NOD mice are characterized by an increased frequency of insulin-containing, smaller-sized, pancreatic islets. Although mice remain diabetes free, florid immune infiltrate remains. However, this immune infiltrate is skewed toward a regulatory phenotype in both T- and B-cell compartments. Pancreatic islets have an increased frequency of IL-10–producing B cells and associated cell surface markers. Resident memory CD69+CD8+ T cells show a significant shift toward reduced CD103 expression, while CD4+ T cells have increased FoxP3+CTLA4+ expression. These data indicate that naturally protected NOD mice have a unique islet signature and provide new insight into regulatory mechanisms within pancreatic islets.

Type 1 diabetes is an organ-specific autoimmune disease characterized by immune-mediated β-cell destruction in pancreatic islets, which results in deficient insulin production. Similar to humans, NOD mice develop spontaneous type 1 diabetes. However, in NOD mouse colonies worldwide, ∼20% (or more) of NOD mice remain normoglycemic and “protected” from diabetes, despite their genetic predisposition (1). Few studies have been done to discover the mechanism of this natural protection. Recently, we have dissected the B-cell functionality in naturally protected NOD mice, highlighting an increased interleukin-10 (IL-10)–producing B-cell frequency and enhanced response to dendritic cells, compared with NOD mice that have developed diabetes (2). Furthermore, it has been suggested that B cells, specifically anergic CD40+IL-10–producing B cells, found in the pancreatic islets of long-term normoglycemic mice (protected) (3), may confer this natural protection. Currently, the phenotype and function of CD4+ and CD8+ T-cell populations in naturally protected NOD mice are unexplored.

Pancreatic islets have a dynamic tissue microenvironment in which immune cells communicate to drive β-cell destruction. This is complicated by cellular and kinetic heterogeneity in both mouse and human pancreatic islets, including the rate of β-cell destruction. The aim of this study was to investigate the characteristics and heterogeneity of the islets in naturally protected NOD mice, including the immune infiltrate. This knowledge may provide insight into disease heterogeneity in humans because not all at-risk individuals develop type 1 diabetes.

Mice

Female NOD/Caj mice, originally from Yale University, were bred in-house at Cardiff University. All mice received water and irradiated food ad libitum and were housed in specific-pathogen-free isolators or scantainers, with a 12-h dark/light cycle, at Cardiff University. All animal experiments were approved by the Cardiff University ethical review process and conducted under U.K. Home Office license in accordance with the U.K. Animals (Scientific Procedures) Act 1986 and associated guidelines. Diabetes conversion rates for Cardiff University can be found in Chen et al. (4), with ∼80% incidence in females by 30 weeks and a median incidence at 19 weeks old. NOD female incidence at other institutions or companies can vary; for example, at The Jackson Laboratory, incidence is ∼90% by 30 weeks of age, with a median female onset at 18 weeks.

Diabetes Incidence

Mice were monitored weekly for glycosuria (Bayer Diastix) from 12 weeks of age. After two positive glycosuria measurements, blood glucose levels were tested, and if >13.9 mmol/L, mice were diagnosed as diabetic. NOD mice that were ≥35 weeks of age and had never tested positive for glycosuria were considered to be protected from diabetes because the incidence of diabetes after this age is very low.

Islet Preparation

Pancreata were inflated with collagenase P solution (1.1 mg/mL) (Roche, Welwyn Garden City, U.K.) in Hanks’ balanced salt solution (with Ca2+ and Mg2+) through the common bile duct, followed by collagenase digestion with shaking at 37°C for 10 min. Islets were isolated by Histopaque density centrifugation (Sigma-Aldrich, Dorset, U.K.) and hand-picked under a dissecting microscope. For flow cytometric analysis, islets were then trypsinized to generate a single-cell suspension. Islet cells were rested at 37°C in 5% CO2 in Iscove’s modified Dulbecco’s media (supplemented with 5% FBS, 2 mmol/L L-glutamine, 100 units/mL penicillin, 100 μg/mL streptomycin, and 50 μmol/L 2-mercaptoethanol) overnight before multiparameter staining.

Fluorescence Immunohistochemistry

Pancreatic tissues were frozen in optimal cutting temperature medium and sectioned at 7-μm thickness. For wholemounts, pancreatic islets were fixed overnight at 4°C in 1% paraformaldehyde. For pancreatic sections, sections were fixed in 1% paraformaldehyde for 1 h at room temperature. Following fixation, tissue was permeabilized with 0.2% Triton X-100 and blocked with 5% FBS before the addition of a rat anti-mouse CD45 (BioLegend, London, U.K.) and a biotinylated anti-insulin (clone D6C4; Abcam) antibody mix. Secondary labeling was performed with both Alexa Fluor 633–conjugated goat anti-rat antibody (Invitrogen, Waltham, MA) and a streptavidin-conjugated Alexa Fluor 488 antibody (Invitrogen) and mounted with VECTASHIELD mounting medium with DAPI (Vector Laboratories). Islet whole-mounts were centrifuged at 300g for 3 min and then resuspended in mounting medium, with DAPI, before mounting to the slide. All sections and whole-mounts were imaged on a Leica SP5 confocal microscope.

ImageJ Analysis

All analyses were performed using Fiji (ImageJ) software (5). Islet area, perimeter, circularity, CD45, and insulin intensity were measured by using a region of interest on individual channels using Fiji’s measurement tool. Islets with insulin remaining were considered to be insulin-containing islets (ICIs) when three or more insulin-positive β-cells were present.

Flow Cytometry

Cells were incubated with TruStain (anti-mouse CD16/32; BioLegend) for 10 min at 4°C followed by fluorochrome-conjugated monoclonal antibodies (mAbs) against cell surface markers for 30 min at 4°C. B-cell phenotyping multiparameter flow cytometry was carried out using the following mAbs: CD3 (145-2C11), B220 (RA3-6B2), CD138 (281-2), CD86 (PO3), CD80 (16-10A1), CD11c (N418), CD11b (M1/70), CD19 (6D5), CD44 (IM7), BAFFR (7H22-E16), MHC class II (10-3-6), and Ki67 (11F6), all purchased from BioLegend. IL-10 (JES5-16E3), IgD (11-26c.2a), and CD40 (3/23) were purchased from BD Biosciences. Galectin-1 antibody was purchased from R&D Systems. T-cell phenotyping multiparameter flow cytometry was carried out using the following mAbs: CD3 (145-2C11), CD8 (53-6.7), CD4 (GK1.5), CD103 (2E7), CD69 (H1-2F3), PD-1 (29F.1A12), interferon-γ (IFN-γ) (XMG1.2), CTLA4 (UC10-4B9), and FoxP3 (MF-14), all purchased from BioLegend. CD25 (PC61) and IL-10 (JES5-16E3) were purchased from BD Biosciences. Dead cells were excluded from analysis by live/dead exclusion dye (Invitrogen). IFN-γ, IL-10, CD107a, and galectin-1 were detected by intracellular cytokine staining after 3 h of stimulation with phorbol myristate acetate (50 ng/mL), ionomycin (500 ng/mL), and monensin (3 µg/mL) (all from Sigma-Aldrich). After extracellular staining, cells were fixed using a fixation/permeabilization kit (BD Biosciences) according to the manufacturer’s instructions and subsequently stained for mAbs against intracellular cytokines or appropriate isotype controls. For FoxP3, CTLA4, and Ki67 staining, cells were fixed/permeabilized using eBioscience nuclear transcription kit. Cell suspensions were acquired on an LSRFortessa (FACSDiva software; BD Biosciences) and analyzed using FlowJo version 10.1 software (Tree Star, Ashland, OR).

Statistical Analysis

Statistical analyses were performed using GraphPad Prism (GraphPad Software, San Diego, CA). Comparison between groups was determined by Mann-Whitney U test or Kolmogorov-Smirnov test. For correlations, Pearson correlation coefficient was calculated. Data were considered significant at P < 0.05.

Data and Resource Availability

The data sets generated or analyzed during the current study are available upon reasonable request.

Increased Frequency of Insulin-Containing Small Pancreatic Islets in Naturally Protected NOD Mice

To investigate the features of the pancreatic islets in naturally protected NOD mice (not diabetic by 35 weeks of age and hereafter referred to as protected), we used immunofluorescence histochemistry, including both pancreatic islet whole-mounts and sections. First, we analyzed size, by area measured in pixels/μm, of pancreatic islets in wholemounts from protected NOD mice (Fig. 1A and B). Representative images (Fig. 1A) and a summary graph (Fig. 1B) demonstrated a range of sizes in the remaining islets in protected NOD mice. Next, we determined the size, by area, of pancreatic islet sections, from both protected and diabetic NOD mice (Fig. 1C and D). Smaller pancreatic islets were significantly more frequent in NOD mice that were protected from diabetes compared with mice that had developed diabetes (P < 0.001) (Fig. 1D). To analyze these islet data further, we used a frequency distribution graph to show the relative contribution, in percentages, of each islet to set “bins” according to islet area. Islet size distribution analysis (in percentages) showed that the relative frequency of islets with an area <50,000 pixels/μm, was greater in protected (85%) than in diabetic (51%) NOD mice (Fig. 1E). Further analysis in both protected and diabetic NOD mice revealed that ICIs with detectable insulin-positive β-cells were significantly smaller in size and more frequent in the protected NOD mouse pancreata compared with diabetic NOD mice (P < 0.01) (Fig. 1F). Interestingly, in the diabetic NOD mice, the very few ICIs detected were larger in area (Fig. 1G). Crucially, a comparison between ICIs and insulin-deficient islets (IDIs) in both NOD groups revealed that ICIs were significantly smaller in islet area (Fig. 1H) and were more frequent (Fig. 1I) compared with IDIs in protected NOD mice. However, this feature was lost in diabetic NOD mice, with no significant difference found in islet area (Fig. 1H) or frequency (Fig. 1I) in the few ICIs identified.

Figure 1

Pancreatic islets from naturally protected NOD mice are smaller in size. Pancreatic islet whole-mounts and sections from naturally protected NOD mice (>35 weeks old) (blue) and NOD mice that had developed diabetes (gray) were analyzed by immunofluorescence staining for CD45 (yellow), insulin (red) and DAPI (blue). A: Representative images of whole-mounts from naturally protected >35-week-old mice. B: Graph showing the distribution of individual islet area in >35-week-old mice. C: Representative images from pancreatic sections from >35-week-old mice and mice that developed diabetes. D and E: Summary graphs of islet area from individual islets in >35-week-old and diabetic NOD mice and the relative frequency distribution in percentage of pancreatic islets. F and G: Summary graphs for ICIs in both >35-week-old and diabetic NOD mice showing area (F) and frequency distribution in the number of islets and relative frequency distribution in percentage (G). H and I: Summary graphs for ICIs and IDIs from >35-week-old and diabetic NOD mice showing islet area (H) and relative frequency distribution of percentage of these islets in diabetic NOD mice and >35-week-old NOD (I). Data are from at least two independent experiments (protected n = 3, diabetic n = 3). *P < 0.05, **P < 0.01, ***P < 0.001 by Kolmogorov-Smirnov test.

Figure 1

Pancreatic islets from naturally protected NOD mice are smaller in size. Pancreatic islet whole-mounts and sections from naturally protected NOD mice (>35 weeks old) (blue) and NOD mice that had developed diabetes (gray) were analyzed by immunofluorescence staining for CD45 (yellow), insulin (red) and DAPI (blue). A: Representative images of whole-mounts from naturally protected >35-week-old mice. B: Graph showing the distribution of individual islet area in >35-week-old mice. C: Representative images from pancreatic sections from >35-week-old mice and mice that developed diabetes. D and E: Summary graphs of islet area from individual islets in >35-week-old and diabetic NOD mice and the relative frequency distribution in percentage of pancreatic islets. F and G: Summary graphs for ICIs in both >35-week-old and diabetic NOD mice showing area (F) and frequency distribution in the number of islets and relative frequency distribution in percentage (G). H and I: Summary graphs for ICIs and IDIs from >35-week-old and diabetic NOD mice showing islet area (H) and relative frequency distribution of percentage of these islets in diabetic NOD mice and >35-week-old NOD (I). Data are from at least two independent experiments (protected n = 3, diabetic n = 3). *P < 0.05, **P < 0.01, ***P < 0.001 by Kolmogorov-Smirnov test.

Close modal

Alongside islet area, islet perimeter was analyzed, and the features observed in protected NOD mice were further confirmed (Supplementary Fig. 1A–C). In addition to islet size, we investigated whether islet circularity was different in protected NOD mice compared with mice that developed diabetes; however, no difference was observed in total islets analyzed (Supplementary Fig. 1D) or between ICIs and IDIs in either the protected or the diabetic NOD mouse cohort (Supplementary Fig. 1E).

Morphological Characterization of Prediabetic NOD Pancreatic Islets

Since protected mice had an increased frequency of smaller islets than diabetic mice, we determined whether smaller islets were less affected by immune cell infiltrate when pancreatic insulitis is established and diabetes begins to manifest (mice aged 12–18 weeks). Whole-mount pancreatic islets were stained with insulin and CD45 (a marker of immune cells) to identify islets undergoing attack, and the size was measured by area. Figure 2A demonstrates heterogeneity of pancreatic islets, shown by representative pictures from two different NOD mice. Z-stacks taken from two individual islets are also shown in Supplementary Fig. 2A. As expected, size, shape, and quantity of the CD45 immune infiltrate were variable in each individual islet. However, we observed a clear pattern between size and quantity of CD45 immune infiltrate. We confirmed this with correlation analysis comparing islet area (Fig. 2B) and islet perimeter (Supplementary Fig. 2B) with fluorescence intensity of CD45 (left) and insulin (right). A significant positive correlation was observed (P < 0.001) between islet area and CD45 immune infiltrate alongside a significant negative correlation (P < 0.01) between islet area and insulin-positive β-cells.

Figure 2

Morphological characterization of prediabetic NOD pancreatic islets. Pancreatic islet whole-mounts from NOD mice aged 12–18 weeks were analyzed by immunofluorescence staining for the immune cell marker CD45 (yellow) and β-cell marker insulin (red). A: Representative images from two individual NOD mice (top and bottom panels); eight pancreatic islets are shown for each mouse. B: Correlative graphs for both CD45 and insulin expression against islet area. Data are from four independent experiments (n = 8).

Figure 2

Morphological characterization of prediabetic NOD pancreatic islets. Pancreatic islet whole-mounts from NOD mice aged 12–18 weeks were analyzed by immunofluorescence staining for the immune cell marker CD45 (yellow) and β-cell marker insulin (red). A: Representative images from two individual NOD mice (top and bottom panels); eight pancreatic islets are shown for each mouse. B: Correlative graphs for both CD45 and insulin expression against islet area. Data are from four independent experiments (n = 8).

Close modal

Regulatory B Cells Are Increased in the Islets of Naturally Protected NOD Mice

Because the pancreatic islets of naturally protected NOD mice have considerable immune infiltration, although remaining normoglycemic, we analyzed the B-cell infiltrate to investigate the frequency of regulatory cells by flow cytometry (Fig. 3). First, we compared the percentage of total B cells in groups of pooled protected NOD mice (>35 weeks) compared with groups of pooled younger prediabetic NOD mice (6–8 weeks old) (Fig. 3A and B) and observed a significant increase in CD19+ B cells (P < 0.001). Second, complementing our previous observation that naturally protected NOD mice have increased splenic IL-10–producing B cells (2), we showed a significant increase in pancreatic islet IL-10–producing B cells (Fig. 3C and Supplementary Fig. 3A). Furthermore, we demonstrated a population of galectin-1–positive B cells (Supplementary Fig. 3B), which encompassed the majority of the IL-10–producing B cells and were increased in >35-week-old mice (P = 0.06) (Fig. 3D and Supplementary Fig. 3C). Interestingly, we also observed a significant increase in CD40+- (Fig. 3E) and CD80+-expressing B cells (Fig. 3F) (for representative gating, see Supplementary Fig. 3D), both markers associated with regulatory B-cell function (6,7). It should be noted that no significant differences were observed in B-cell expression of MHC class II, CD86, or BAFFR (Supplementary Fig. 3E and F).

Figure 3

Naturally protected NOD mice have increased regulatory B cells in the pancreatic islet infiltrate. Pancreata from groups (n = 2–3) of NOD mice aged 6–8 weeks (younger prediabetic) (red) and >35 weeks (protected) (blue) were taken, and pancreatic islets pooled together before flow cytometric analysis. A: Representative histograms showing CD19+ B-cell expression. B: Overall percentages for CD19+ B cells. CF: Overall percentages of CD19+ B cells expressing IL-10 (C), IL-10+ in galectin-1–positive and –negative (Gal-1+ and Gal-1) compartments (D), CD40 (E), and CD80 (F). GI: Representative flow cytometry plots gated on live CD3CD11bCD11c cells for CD138 and IgD populations (CD138IgD+ [blue gate], CD138+IgD+ [orange gate], CD138+IgD [gray gate], CD138hiIgD [red gate]) (G), CD19 expression on each of the CD138/IgD populations (H), and histograms showing CD44 and Ki67 on each of the CD138/IgD populations (I). J and K: Overall percentages for CD138+IgD+ cells (J) and CD138IgD+ cells (K). L: Ratio of CD138+IgD+ to CD138IgD+ B cells. M: Overall percentages of CD138+IgD cells. N: Ratio of CD138+IgD cells to CD138IgD+ B cells. O: Summary graph for percentages of CD138hiIgD plasmablasts in both young NOD and protected >35-week-old NOD mouse groups. Data are from at least four independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001 by Mann-Whitney U test.

Figure 3

Naturally protected NOD mice have increased regulatory B cells in the pancreatic islet infiltrate. Pancreata from groups (n = 2–3) of NOD mice aged 6–8 weeks (younger prediabetic) (red) and >35 weeks (protected) (blue) were taken, and pancreatic islets pooled together before flow cytometric analysis. A: Representative histograms showing CD19+ B-cell expression. B: Overall percentages for CD19+ B cells. CF: Overall percentages of CD19+ B cells expressing IL-10 (C), IL-10+ in galectin-1–positive and –negative (Gal-1+ and Gal-1) compartments (D), CD40 (E), and CD80 (F). GI: Representative flow cytometry plots gated on live CD3CD11bCD11c cells for CD138 and IgD populations (CD138IgD+ [blue gate], CD138+IgD+ [orange gate], CD138+IgD [gray gate], CD138hiIgD [red gate]) (G), CD19 expression on each of the CD138/IgD populations (H), and histograms showing CD44 and Ki67 on each of the CD138/IgD populations (I). J and K: Overall percentages for CD138+IgD+ cells (J) and CD138IgD+ cells (K). L: Ratio of CD138+IgD+ to CD138IgD+ B cells. M: Overall percentages of CD138+IgD cells. N: Ratio of CD138+IgD cells to CD138IgD+ B cells. O: Summary graph for percentages of CD138hiIgD plasmablasts in both young NOD and protected >35-week-old NOD mouse groups. Data are from at least four independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001 by Mann-Whitney U test.

Close modal

Many B cells in pancreatic islets express CD138 (8,9), a marker that identifies plasmablasts and plasma cells. We investigated our previously described populations identified by CD138 and IgD expression (9), examining them further (Fig. 3G–I) using markers for murine plasmablasts (10,11). We assessed CD19 (Fig. 3H), CD44, and Ki67 (Fig. 3I) in each CD138+/− population, revealing that CD138hiIgD cells (red gate) contained a CD44hiKi67+ highly proliferative population that also expressed intermediate levels of CD19 (CD19int). With this further analysis, we propose that CD138hiIgD cells (red gate) are a subpopulation of dividing plasmablasts. Few B cells that remained IgD+ were proliferating, and so it is probable that they represent classical B cells (CD138IgD+) (blue gate) and an intermediate stage of plasmablast (CD138+IgD+) (orange gate). CD138+IgD cells (gray gate) were a mixture of both CD19+/− cells and increased Ki67 expression, compared with CD138IgD+ classical B cells, and are likely to represent both nondividing intermediate-stage plasmablasts and dividing plasmablasts.

We demonstrated that protected NOD mice displayed significant increases in both CD138+IgD+ B cells (Fig. 3J) and CD138IgD+ B cells (Fig. 3K) compared with young, 6–8-week-old mice. This observation indicated that CD138+IgD+ cells were not selectively recruited over CD138IgD+ B cells (Fig. 3L). However, the frequency of CD138+IgD B cells in both young and older protected NOD mouse groups was not altered (Fig. 3M). Thus, the ratio of the CD138+IgD to CD138IgD+ B cells was increased in the younger NOD mice (Fig. 3N). This may suggest that these B cells arrive early to the pancreatic islets. Analysis of the small population of dividing plasmablasts (CD138hiIgD) showed a significant increase in protected NOD mice compared with younger NOD mice (Fig. 3O).

Enrichment of CD4+FoxP3+CTLA4+ Regulatory T Cells in Naturally Protected NOD Mice

To determine the characteristics of CD4+ T cells in the pancreatic islets of NOD mice, that are naturally protected from diabetes, we studied the islet-infiltrating T cells of groups of pooled protected NOD mice by multiparameter flow cytometry and compared these to islet-infiltrating T cells from groups of pooled mice 6–8 weeks of age (mice with early-stage insulitis). CD4+ T-cell frequency and expression were increased in protected NOD mice, although nonsignificantly because of the variability in younger NOD mice (Fig. 4A and B). To ascertain whether CD4+ regulatory T cells (Tregs) contributed to the protection seen in protected NOD mice, we investigated the presence of CD4+FoxP3+ Tregs in the pancreatic islets and revealed a significant increase in CD4+FoxP3+ Tregs in the pancreatic islets of protected NOD mice compared with 6–8-week-old mice (Fig. 4C and D). Further analysis of the CD4+FoxP3+ T cells revealed a significant increase in the frequency of CTLA4+CD4+FoxP3+ T cells in protected NOD mice compared with 6–8-week-old mice (P < 0.01) (Fig. 4E and F). No significant differences were observed in the percentage of CD4+FoxP3+ T cells expressing CD25, PD-1, or CD103. Interestingly, intracellular cytokine analysis of both CD4+FoxP3−/+ T cells showed a significant increase in IFN-γ–producing CD4+FoxP3 T cells in protected NOD mice compared with mice aged 6–8 weeks (Fig. 4G and H). No differences in IL-10 in FoxP3+CD4+ T cells were observed. This increase in IFN-γ production from restimulated CD4+ T cells may reflect enhanced antigen experience.

Figure 4

Increased frequency of CD4+CTLA4+FoxP3+ Tregs in naturally protected NOD mice. Pancreata from groups (n = 2–3) of NOD mice aged >35 weeks (protected) (blue) and 6–8 weeks (younger prediabetic) (red) were taken, and pancreatic islets pooled together before flow cytometric analysis. A: Representative histogram of CD4 expression. B: CD4+ T-cell percentages and CD4 geometric mean fluorescence intensity (GMFI). C and D: CD4+FoxP3+ T-cell expression with representative histograms (C) and summary graph (D). E and F: Expression of CD4+ Treg markers on CD4+FoxP3+ and CD4+FoxP3 T cells with representative histograms for CTLA4, CD25, PD-1, and CD103 (E) and graphs showing summary percentages (F). G and H: IFN-γ and IL-10 expression in CD4+FoxP3+/− subsets with representative flow plots (G) and summary graphs (H). CD4+ T cells were gated on single, live cells. Data are from at least three independent experiments. *P < 0.05, **P < 0.01 by Mann-Whitney U test.

Figure 4

Increased frequency of CD4+CTLA4+FoxP3+ Tregs in naturally protected NOD mice. Pancreata from groups (n = 2–3) of NOD mice aged >35 weeks (protected) (blue) and 6–8 weeks (younger prediabetic) (red) were taken, and pancreatic islets pooled together before flow cytometric analysis. A: Representative histogram of CD4 expression. B: CD4+ T-cell percentages and CD4 geometric mean fluorescence intensity (GMFI). C and D: CD4+FoxP3+ T-cell expression with representative histograms (C) and summary graph (D). E and F: Expression of CD4+ Treg markers on CD4+FoxP3+ and CD4+FoxP3 T cells with representative histograms for CTLA4, CD25, PD-1, and CD103 (E) and graphs showing summary percentages (F). G and H: IFN-γ and IL-10 expression in CD4+FoxP3+/− subsets with representative flow plots (G) and summary graphs (H). CD4+ T cells were gated on single, live cells. Data are from at least three independent experiments. *P < 0.05, **P < 0.01 by Mann-Whitney U test.

Close modal

Islet CD8 Resident Memory T Cells in Naturally Protected NOD Mice Switch to a CD103 Phenotype

Additional analysis of T cells was performed. Similar to CD4+ T cells, CD8+ T cells were modestly increased in frequency in >35-week-old mice (Fig. 5A and B). Because CD8+ T cells are found from an early age in the pancreatic tissue, we investigated markers of tissue residency. CD8+ tissue resident memory cells (TRM) can be distinguished by the surface markers CD69 and CD103 (12). CD8+ TRM populations have now been widely studied in various tissues and play a crucial role in immunosurveillance and protect against secondary viral infections (13). We demonstrated three key populations of CD8+ T cells in the pancreas of NOD mice: CD103CD69 recirculating cells, CD103CD69+ TRM, and CD103+CD69+ TRM (Fig. 5D–G). Protected >35-week-old NOD mice and younger NOD mice had similar frequencies of recirculating CD8+CD103CD69 T cells (Fig. 5D). Strikingly, CD8+ TRM were significantly different between protected NOD mice and mice aged 6–8 weeks, with a shift toward greater prominence of CD103 TRM in protected NOD mice (Fig. 5E and F). Furthermore, both CD8+CD103−/+ TRM populations expressed CD107a, IFN-γ, and PD-1, with the CD103 TRM becoming more activated overall, after stimulation (Fig. 5G). There was no enrichment of PD-1 (a marker for T-cell exhaustion [14]) in either TRM population (Fig. 5G). Further analysis of the CD107a+ cells (a marker for recent degranulation) in the TRM populations revealed that CD107a+CD103+ TRM had fewer IFN-γ+PD-1-expressing cells than the CD107a+CD103 TRM (P < 0.05) (Fig. 5H, summary graph) but, overall, a greater proportion of PD-1 expression (Fig. 5I, pie charts). Evaluation of IFN-γ and PD-1 subpopulations in the younger as well as the protected NOD mice showed a significant increase in IFN-γ+PD-1+ T cells in both CD103+/− TRM populations (Fig. 5J and K). Overall, naturally protected NOD mice showed a shift toward a CD8+CD103 TRM population, which has a more activated phenotype after stimulation, alongside an increase in IFN-γ in the TRM subsets.

Figure 5

CD8+ TRM in naturally protected NOD mice switch to a CD103 phenotype. Pancreatic islets from groups (n = 2–3) of NOD mice aged >35 weeks (protected) (blue) and 6–8 weeks (younger prediabetic) (red) were taken and pancreatic islets pooled together before flow cytometric analysis. A: Representative flow plots showing CD8+ T cells. B: CD8+ T-cell percentages. C: Representative flow plots to show CD103CD69 recirculating CD8+ T cells and CD103+/−CD69+ TRM. DF: Percentages of CD8+ T-cell populations demonstrating CD103CD69 recirculating T cells (D), CD103CD69+ TRM (E), and CD103+CD69+ TRM (F). G: Pie charts summarizing total percentages of CD107a, IFN-γ and PD-1 expression on both CD103+/− TRM CD8+ subsets. H and I: CD8+CD103+/− TRM subset analysis gated on CD107+ cells containing IFN-γ+PD-1, IFN-γ+PD-1+, and IFN-γPD-1+ subsets, showing overall percentages of IFN-γ and PD-1 populations (H) and illustrating (with pie charts) the proportion of CD107a-expressing cells for IFN-γ and PD-1 populations (I). J and K: IFN-γ+ and PD-1+ populations on the CD107a+ subset of CD8+CD103CD69+ TRM (top) and CD8+CD103+CD69+ TRM (bottom) from 6–8-week-old (red) and >35-week-old (blue) NOD mice, showing representative flow cytometry plots (J) and summary graphs of percentages (K). CD8+ T cells were gated on single, live cells. Data are from at least three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001 by Mann-Whitney U test.

Figure 5

CD8+ TRM in naturally protected NOD mice switch to a CD103 phenotype. Pancreatic islets from groups (n = 2–3) of NOD mice aged >35 weeks (protected) (blue) and 6–8 weeks (younger prediabetic) (red) were taken and pancreatic islets pooled together before flow cytometric analysis. A: Representative flow plots showing CD8+ T cells. B: CD8+ T-cell percentages. C: Representative flow plots to show CD103CD69 recirculating CD8+ T cells and CD103+/−CD69+ TRM. DF: Percentages of CD8+ T-cell populations demonstrating CD103CD69 recirculating T cells (D), CD103CD69+ TRM (E), and CD103+CD69+ TRM (F). G: Pie charts summarizing total percentages of CD107a, IFN-γ and PD-1 expression on both CD103+/− TRM CD8+ subsets. H and I: CD8+CD103+/− TRM subset analysis gated on CD107+ cells containing IFN-γ+PD-1, IFN-γ+PD-1+, and IFN-γPD-1+ subsets, showing overall percentages of IFN-γ and PD-1 populations (H) and illustrating (with pie charts) the proportion of CD107a-expressing cells for IFN-γ and PD-1 populations (I). J and K: IFN-γ+ and PD-1+ populations on the CD107a+ subset of CD8+CD103CD69+ TRM (top) and CD8+CD103+CD69+ TRM (bottom) from 6–8-week-old (red) and >35-week-old (blue) NOD mice, showing representative flow cytometry plots (J) and summary graphs of percentages (K). CD8+ T cells were gated on single, live cells. Data are from at least three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001 by Mann-Whitney U test.

Close modal

In this report, we demonstrate key characteristics of pancreatic islets in a group of mice that are naturally protected from developing spontaneous diabetes. First, we discovered that smaller islets remain in these protected mice, with a clear correlation between islet size and immune infiltrate. Furthermore, insulin-positive β-cells are still present in pancreatic islets despite florid immune infiltrate. This immune infiltrate has a high frequency of B and T cells; however, the compositional signature was notably different in both immune cell compartments.

For the first time, we show that protected NOD mice have an increased frequency of smaller islets remaining in the pancreas, with ICIs smaller in size compared with IDIs. NOD mice that developed diabetes did not display this pattern. Moreover, we show in prediabetic NOD mice that larger islets have larger immune infiltrates. Previously, islet size has been shown to decrease as the duration of disease progresses in the NOD mouse (15); however, more sophisticated imaging identified that the smaller islets, located peripherally, are destroyed earlier in the disease process (16). However, surprisingly, the CD3+ immune infiltrate was not localized to the smaller islets or within a particular islet region (16). In humans, individuals with recent-onset type 1 diabetes have larger islets compared with people with long-standing type 1 diabetes (17).

Islets smaller in size than those found in the 12–16-week-old NOD mice may have been destroyed; however, we noted that islet size and infiltrate were correlated, but curiously, the very few remaining islets with insulin-positive β-cells were not smaller in size. Certainly, no correlation between β-cell mass and insulitis was observed in human pancreatic sections from donors with type 1 diabetes, but islet size was not addressed (18). This dichotomy requires further study to ascertain why this is the case. An explanation for this correlation between islet size and immune infiltrate could be explained by an increased capillary density (19), providing more immune cell access.

IL-10–producing B cells diminish the inflammatory response (20). Building on previous work (2), we now show that naturally protected NOD mice have a regulatory B-cell bias in the pancreatic islets. We demonstrate that galectin-1–positive B cells in pancreatic islets, a marker shown to be necessary for the function of regulatory B cells (21), and production of this protein, from activated B cells, can influence T-cell responses, including inducing T-cell apoptosis (22). Furthermore, the infiltrating B cells have increased levels of other cell surface markers associated with regulation. First, the CD80 molecule is known to preferentially bind CTLA4 (23), which we also found to be significantly increased on CD4+FoxP3+ Tregs in these protected NOD islets. Second, IL-10–producing B cells require CD40 to suppress effector T cells and autoimmunity (6). Interestingly, we describe here a significant increase in dividing CD138hiCD44hi plasmablasts in protected NOD mice previously reported to be an IL-10–producing population (10).

We observed that CD138+IgD pancreatic islet B cells are similar in frequency in both younger and protected NOD mice. Early islet B cells have a B1 B-cell phenotype (24), B cells that are preferentially located in the peritoneum and pleural cavities, and are players in the initiation of type 1 diabetes (25). CD138+IgD cells identified here are a heterogenous pool of dividing and nondividing plasmablasts at various intermediate differentiation stages, which consist of CD19+ and CD19 cells that lack IgM expression and are more proliferative compared with classical B cells. Crucially, this subset also contains antigen-specific B cells (9). Further work on these defined subsets is required to understand whether they are a B1-like cell that has altered as a result of the inflamed tissue environment.

Finally, we identified a significant shift in the CD8+ T-cell compartment. A CD8+CD103+ TRM phenotype is more prevalent in younger NOD mice, whereas a CD8+CD103 TRM phenotype dominates in protected NOD mice, with CD8+CD103 TRM producing increased IFN-γ when restimulated ex vivo. CD8+ TRM respond rapidly to control local immune responses in the tissue and so are central in tissue immunosurveillance. CD8+CD103 TRM have been described in various tissues, with CD8+CD103+ TRM located preferentially in the epidermis while CD8+CD103 TRM are located in the dermis of human skin (26), with distinct functional roles (26,27). CD8+CD103+/− TRM have been defined as separate populations, with different patterns of recall, molecular signature, and migration (2729). CD8+CD103 TRM are more transient in the tissue compared with CD103+ counterparts (28), alongside an increased expression of sphingosine-1-phosphate receptor 1 (27). It remains unclear whether the enrichment of the CD8+CD103 TRM population in the older protected mice represents a more transient CD8 TRM population, and this would require further interrogation. CD8+ TRM in human pancreatic tissue from adults with recent-onset type 1 diabetes have been identified, but interestingly, only CD8+CD103+ TRM were detected in pancreatic tissue (30). However, in donors without diabetes, a CD8+CD103 TRM phenotype has been observed in ∼20–30% of resident CD8+ T cells in pancreatic islets (31).

E-cadherin, the principal ligand for CD103, is expressed by pancreatic islets (32), and this interaction can result in the release of cytokines and lytic granules from CD8+ T cells, therefore controlling cytotoxic CD8+ T-cell responses (33,34). CD103 is also required for the efficient destruction of pancreatic islet allografts (35). A shift toward CD8+ T cells lacking CD103 in naturally protected NOD mice may result in reduced targeted cell death. However, this CD103 TRM shift may also be a result from the loss of the ligand E-cadherin because of substantial loss of pancreatic islets.

In conclusion, naturally protected NOD mice have a unique pancreatic signature, with remaining islets that contain smaller insulin-producing β-cells and an immune infiltrate (both T and B cells) shifted toward a regulatory phenotype. These results are important to understanding the balance between a destroyed islet and an islet that remains, even partially, intact. Limitations in this study reflect the difficulty of studying both heterogenous islets and immune cell subsets, especially with limited cell numbers. Furthermore, protected NOD mice represent a pool of mice that cannot be detected early in the disease process, and so, comparison with mice that are younger, including mice that have established insulitis, is challenging. These younger mice would encompass both those that will develop diabetes and those that will be naturally protected, and we cannot, as yet, predict which mice will develop diabetes and distinguish them from mice that will be spared. Nevertheless, these observations highlight the need for further investigation into the dynamic process of β-cell destruction.

J.B. is currently affiliated with the Institute of Biomedical & Clinical Science, University of Exeter, Exeter, U.K.

This article contains supplementary material online at https://doi.org/10.2337/figshare.13643393.

Funding. This work was funded by Medical Research Council (U.K.) grant MR/K021141/1 to F.S.W.

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. J.B. performed the experiments and analyzed the data. J.B. and F.S.W. designed the experiments and wrote the manuscript. T.C.T. and J.D. contributed to the experimental procedures. All authors reviewed the manuscript. F.S.W. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

1.
Anderson
MS
,
Bluestone
JA
.
The NOD mouse: a model of immune dysregulation
.
Annu Rev Immunol
2005
;
23
:
447
485
2.
Boldison
J
,
Da Rosa
LC
,
Davies
J
,
Wen
L
,
Wong
FS
.
Dendritic cells license regulatory B cells to produce IL-10 and mediate suppression of antigen-specific CD8 T cells
.
Cell Mol Immunol
2020
;
17
:
843
855
3.
Kleffel
S
,
Vergani
A
,
Tezza
S
, et al
.
Interleukin-10+ regulatory B cells arise within antigen-experienced CD40+ B cells to maintain tolerance to islet autoantigens
.
Diabetes
2015
;
64
:
158
171
4.
Chen
D
,
Thayer
TC
,
Wen
L
,
Wong
FS
.
Mouse models of autoimmune diabetes: the Nonobese Diabetic (NOD) mouse
.
Methods Mol Biol
2020
;
2128
:
87
92
5.
Schindelin
J
,
Arganda-Carreras
I
,
Frise
E
, et al
.
Fiji: an open-source platform for biological-image analysis
.
Nat Methods
2012
;
9
:
676
682
6.
Yoshizaki
A
,
Miyagaki
T
,
DiLillo
DJ
, et al
.
Regulatory B cells control T-cell autoimmunity through IL-21-dependent cognate interactions
.
Nature
2012
;
491
:
264
268
7.
Suvas
S
,
Singh
V
,
Sahdev
S
,
Vohra
H
,
Agrewala
JN
.
Distinct role of CD80 and CD86 in the regulation of the activation of B cell and B cell lymphoma
.
J Biol Chem
2002
;
277
:
7766
7775
8.
Serreze
DV
,
Chapman
HD
,
Niens
M
, et al
.
Loss of intra-islet CD20 expression may complicate efficacy of B-cell-directed type 1 diabetes therapies
.
Diabetes
2011
;
60
:
2914
2921
9.
Boldison
J
,
Da Rosa
LC
,
Buckingham
L
,
Davies
J
,
Wen
L
,
Wong
FS
.
Phenotypically distinct anti-insulin B cells repopulate pancreatic islets after anti-CD20 treatment in NOD mice
.
Diabetologia
2019
;
62
:
2052
2065
10.
Matsumoto
M
,
Baba
A
,
Yokota
T
, et al
.
Interleukin-10-producing plasmablasts exert regulatory function in autoimmune inflammation
.
Immunity
2014
;
41
:
1040
1051
11.
Pracht
K
,
Meinzinger
J
,
Daum
P
, et al
.
A new staining protocol for detection of murine antibody-secreting plasma cell subsets by flow cytometry
.
Eur J Immunol
2017
;
47
:
1389
1392
12.
Carbone
FR
.
Tissue-resident memory T cells and fixed immune surveillance in nonlymphoid organs
.
J Immunol
2015
;
195
:
17
22
13.
Park
CO
,
Kupper
TS
.
The emerging role of resident memory T cells in protective immunity and inflammatory disease
.
Nat Med
2015
;
21
:
688
697
14.
Day
CL
,
Kaufmann
DE
,
Kiepiela
P
, et al
.
PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression
.
Nature
2006
;
443
:
350
354
15.
Novikova
L
,
Smirnova
IV
,
Rawal
S
,
Dotson
AL
,
Benedict
SH
,
Stehno-Bittel
L
.
Variations in rodent models of type 1 diabetes: islet morphology
.
J Diabetes Res
2013
;
2013
:
965832
16.
Alanentalo
T
,
Hörnblad
A
,
Mayans
S
, et al
.
Quantification and three-dimensional imaging of the insulitis-induced destruction of beta-cells in murine type 1 diabetes
.
Diabetes
2010
;
59
:
1756
1764
17.
Seiron
P
,
Wiberg
A
,
Kuric
E
, et al
.
Characterisation of the endocrine pancreas in type 1 diabetes: islet size is maintained but islet number is markedly reduced
.
J Pathol Clin Res
2019
;
5
:
248
255
18.
Campbell-Thompson
M
,
Fu
A
,
Kaddis
JS
, et al
.
Insulitis and β-cell mass in the natural history of type 1 diabetes
.
Diabetes
2016
;
65
:
719
731
19.
Fowler
JL
,
Lee
SS
,
Wesner
ZC
,
Olehnik
SK
,
Kron
SJ
,
Hara
M
.
Three-dimensional analysis of the human pancreas
.
Endocrinology
2018
;
159
:
1393
1400
20.
Fillatreau
S
,
Sweenie
CH
,
McGeachy
MJ
,
Gray
D
,
Anderton
SM
.
B cells regulate autoimmunity by provision of IL-10
.
Nat Immunol
2002
;
3
:
944
950
21.
Alhabbab
R
,
Blair
P
,
Smyth
LA
, et al
.
Galectin-1 is required for the regulatory function of B cells
.
Sci Rep
2018
;
8
:
2725
22.
Zuñiga
E
,
Rabinovich
GA
,
Iglesias
MM
,
Gruppi
A
.
Regulated expression of galectin-1 during B-cell activation and implications for T-cell apoptosis
.
J Leukoc Biol
2001
;
70
:
73
79
23.
Pentcheva-Hoang
T
,
Egen
JG
,
Wojnoonski
K
,
Allison
JP
.
B7-1 and B7-2 selectively recruit CTLA-4 and CD28 to the immunological synapse
.
Immunity
2004
;
21
:
401
413
24.
Ryan
GA
,
Wang
CJ
,
Chamberlain
JL
, et al
.
B1 cells promote pancreas infiltration by autoreactive T cells
.
J Immunol
2010
;
185
:
2800
2807
25.
Diana
J
,
Simoni
Y
,
Furio
L
, et al
.
Crosstalk between neutrophils, B-1a cells and plasmacytoid dendritic cells initiates autoimmune diabetes
.
Nat Med
2013
;
19
:
65
73
26.
Watanabe
R
,
Gehad
A
,
Yang
C
, et al
.
Human skin is protected by four functionally and phenotypically discrete populations of resident and recirculating memory T cells
.
Sci Transl Med
2015
;
7
:
279ra39
27.
Wakim
LM
,
Woodward-Davis
A
,
Liu
R
, et al
.
The molecular signature of tissue resident memory CD8 T cells isolated from the brain
.
J Immunol
2012
;
189
:
3462
3471
28.
Gebhardt
T
,
Whitney
PG
,
Zaid
A
, et al
.
Different patterns of peripheral migration by memory CD4+ and CD8+ T cells
.
Nature
2011
;
477
:
216
219
29.
Wakim
LM
,
Woodward-Davis
A
,
Bevan
MJ
.
Memory T cells persisting within the brain after local infection show functional adaptations to their tissue of residence
.
Proc Natl Acad Sci U S A
2010
;
107
:
17872
17879
30.
Kuric
E
,
Seiron
P
,
Krogvold
L
, et al
.
Demonstration of tissue resident memory CD8 T cells in insulitic lesions in adult patients with recent-onset type 1 diabetes
.
Am J Pathol
2017
;
187
:
581
588
31.
Radenkovic
M
,
Uvebrant
K
,
Skog
O
, et al
.
Characterization of resident lymphocytes in human pancreatic islets
.
Clin Exp Immunol
2017
;
187
:
418
427
32.
Cirulli
V
,
Baetens
D
,
Rutishauser
U
,
Halban
PA
,
Orci
L
,
Rouiller
DG
.
Expression of neural cell adhesion molecule (N-CAM) in rat islets and its role in islet cell type segregation
.
J Cell Sci
1994
;
107
:
1429
1436
33.
Le Floc’h
A
,
Jalil
A
,
Vergnon
I
, et al
.
Alpha E beta 7 integrin interaction with E-cadherin promotes antitumor CTL activity by triggering lytic granule polarization and exocytosis
.
J Exp Med
2007
;
204
:
559
570
34.
Franciszkiewicz
K
,
Le Floc’h
A
,
Boutet
M
,
Vergnon
I
,
Schmitt
A
,
Mami-Chouaib
F
.
CD103 or LFA-1 engagement at the immune synapse between cytotoxic T cells and tumor cells promotes maturation and regulates T-cell effector functions
.
Cancer Res
2013
;
73
:
617
628
35.
Feng
Y
,
Wang
D
,
Yuan
R
,
Parker
CM
,
Farber
DL
,
Hadley
GA
.
CD103 expression is required for destruction of pancreatic islet allografts by CD8(+) T cells
.
J Exp Med
2002
;
196
:
877
886
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/content/license.