B-cells participate in the autoimmune response that precedes the onset of type 1 diabetes, but how these cells contribute to disease progression is unclear. In this study, we analyzed the phenotype and functional characteristics of islet-infiltrating B-cells in the diabetes-prone NOD mouse and in the insulitis-prone but diabetes-resistant (NOD×NOR)F1 mouse. The results indicate that B-cells accumulate in the islets of both mice influenced by sex traits. Phenotypically and functionally, these B-cells are highly affected by the islet inflammatory milieu, which may keep them in a silenced status. Moreover, although islet-infiltrating B-cells seem to be antigen experienced, they can only induce islet-infiltrating T-cell proliferation when they act as accessory cells. Thus, these results strongly suggest that islet-infiltrating B-cells do not activate islet-infiltrating T-cells in situ, although they may affect the progression of the disease otherwise.

Type 1 diabetes develops following the selective loss of pancreatic β-cells due to a self-destructive mechanism mediated by the patient's own immune system (1). The NOD (nonobese diabetic) mouse develops a form of autoimmune diabetes that resembles human type 1 diabetes (2). Studies with this animal model have established that islet-infiltrating β-cell–reactive T-cells are the major effectors of β-cell damage. However, other immune system cells are also crucial in the disease development. Among these cells, B-cells are essential in the onset and progression of type 1 diabetes (rev. in 3), and although it is not fully understood when and how these cells participate in type 1 diabetes, it is known that they produce autoantibodies against many β-cell autoantigens (rev. in 4) and act as antigen-presenting cells (APCs) (3). On the one hand, the production of specific autoantibodies directly correlates with the progression of type 1 diabetes in both humans and NOD mice (5,6). In addition, in NOD mice, the transmission of maternal islet β-cell–reactive autoantibodies during pregnancy contributes to the disease progression in offspring (7). On the other hand, as APCs, B-cells may be involved in regulating the activation of islet-reactive T-cells. This activity may be more relevant in the genesis and/or development of type 1 diabetes, since an impaired B-cell–mediated antigen presentation by major histocompatibility complex class II results in a resistance to spontaneous type 1 diabetes development in NOD mice (810). B-cells may also have a tolerogenic function, as the transfer of activated B-cells prevents type 1 diabetes, probably by triggering the apoptosis of β-cell–reactive cytotoxic T-cells (CTLs) and/or inhibiting APC activity in NOD mice (11). In fact, the antigenic repertoire and functional status of B-cells may be different depending on the anatomical region and stage of the disease, and the behavior and role during the progression of type 1 diabetes may also be different.

B-cells migrate to the pancreatic islets and infiltrate them in the same way as do T-cells, but the reasons remain unknown; however, since most β-cell damage occurs by an in situ CTL response, these islet-infiltrating B-cells might also be relevant in the development of the disease. The aim of this study was to discover the phenotypical and functional traits of islet-infiltrating B-cells in NOD and (NOD×NOR)F1 mice (12). The results indicate that islet-infiltrating B-cells accumulate in the islets influenced by sex traits, and their phenotype is compatible with a downregulated population. Moreover, although islet-infiltrating B-cells seem to be antigen experienced, they can only induce islet-infiltrating T-cell proliferation when they act as accessory cells. In conclusion, islet-infiltrating B-cells may not activate islet-infiltrating T-cells in situ but may contribute to the onset of type 1 diabetes otherwise.

Breeding pairs of NOD, NOR (13), and BALB/c mice were purchased from The Jackson Laboratory (Bar Harbor, ME) and maintained by brother-sister mating under specific pathogen-free conditions, following the European Laws and Directives concerning the protection of animals for experimentation. In our facility, 85% female and 37% male NOD mice developed spontaneous autoimmune diabetes at 32 weeks of age. (NOD×NOR)F1 mice were generated by crossing NOD with NOR mice. Most (NOD×NOR)F1 mice developed a nondestructive but severe form of insulitis, although only ∼19% of (NOD× NOR)F1 female mice developed overt diabetes. We used the group of (NOD× NOR)F1 mice as a resistance-to-disease-progression model to study the characteristics of islet-infiltrating B-cells in nondestructive insulitis.

Tissue immunofluorescence stainings.

Pancreatic cryosections from NOD female mice were air-dried for 30 min and incubated at 4°C with anti-CD45R/B220 (biotin RA3-6B2; BD Biosciences) and then with streptavidin–fluorescein isothiocyanate (FITC) (BD Biosciences) at 4°C for 45 min. After washing, the pancreata sections were stained for 2 h at 4°C with a rabbit anti-Ki67 antibody (NCL-ki67p; Novocastra, Newcastle upon Tyne, U.K.), washed again, and then incubated with the secondary antibody (goat anti-rabbit TRITC; Southern Biotechnology Associates, Birmingham, AL) at 4°C for 45 min.

Antibodies and flow cytometry.

For immunofluorescence staining, islet-infiltrating mononuclear cells were obtained as previously described elsewhere (14). Briefly, after their isolation by enzymatic digestion of the pancreas (Collagenase CLS-4; Worthington Biochemical, Lakewood, NJ), pancreatic islets were handpicked and mechanically disrupted to obtain islet single-cell suspension. Spleens were disrupted into single-cell suspensions and erythrocytes lysed in 0.87% ammonium chloride. B-cell phenotypic analysis was performed by staining the previous samples at 4°C for 20 min with a combination of the following monoclonal antibodies (mAbs): anti-CD45R/B220 (FITC RA3-6B2), anti-CD19 (PE 1D3), anti-CD21 (FITC 7G6), anti-CD5 (FITC 53-7.3), anti-IgA (biotin C10-1), anti-IgD (FITC 11-26c.2a), anti-IgG1 (biotin A85-1), anti-IgG2a2b (biotin R2-40, which also recognizes NOD IgG2c), IgG2b (biotin R12-3), anti-IgG3 (biotin R40-82), anti-IgM (biotin R6-60.2), anti-CD24 (FITC M1/69), anti-CD86 (biotin GL1), anti-CD80 (biotin 16-10A1), anti-CD40 (biotin 3/23), anti-CD44 (biotin IM7), anti-CD69 (FITC H1.2F3), anti–I-Ak (biotin 10-3.6), and anti–H-2kd (biotin SF1-1.1), all of which were purchased from BD Biosciences, and anti-CD11b (biotin M1/70), purchased from Southern Biotechnology Associates. Biotin-conjugated mAbs were subsequently stained with streptavidin-PerCP (BD Biosciences) and analyzed by FACScan Cell Analyzer (BD Biosciences) using CellQuest software. According to their forward and side characteristics, live cells (previously determined by propidium iodide) were gated and analyzed for the expression of the surface molecules of interest. When the fluorescence histogram or any label clearly showed a single peak in the B-cell population, we measured the median fluorescence intensity because this parameter is more constant when analyzing small samples. When the histogram showed a continuous shoulder or a bimodal distribution, we measured the percentage of positive cells. To discard any possible effect of collagenase on membrane cell markers, samples of splenic cells were overdigested with this enzyme at 37°C for 45 min (for islet isolation, we usually perform pancreatic digestion with collagenase at 37°C for 20 min). Samples were then stained and analyzed by flow cytometry, and the results were compared with those obtained from undigested samples.

B-cell stimulation assays.

After enzymatic isolation, pancreatic islets from 12-week female NOD or (NOD×NOR)F1 mice were cultured in 96-well tissue culture plates (20 islets/well) at 37°C in 5% CO2 in complete culture medium (RPMI-1640 media containing 10% heat-inactivated fetal bovine serum [Life Technologies, Grand Island, NY], 100 units/ml penicillin, 100 μg/ml streptomycin, 2 mmol/l glutamine, 1 mmol/l sodium pyruvate, and 25 μmol/l β-mercaptoethanol). Likewise, 2.5 × 105/well splenocytes were cultured under the same conditions. Activating stimuli consisted of 10 μg/ml lipopolysaccharide (LPS) (Sigma-Aldrich, St. Louis, MO) or 10 μg/ml anti-CD40 (clone 3/23; BD Biosciences) plus 10 units/ml rIL-4 (R&D Systems, Minneapolis, MN). Then cultured mononuclear cells, from either spleen or islet infiltrate, were harvested at 0, 24, and 48 h and analyzed by three-color flow cytometry, as previously described.

Cytokine profile analysis.

Splenic and islet-infiltrating mononuclear cells from 12-week NOD and (NOD×NOR)F1 female mice (right after islet isolation) were cultured at 37°C in 5% CO2 for 5 h in complete culture medium in 5 ng/ml phorbol myristic acid and 500 ng/ml Ionomycin (both from Sigma-Aldrich) and 10 μmol/l monensin (eBiosience, San Diego, CA). Then cells were stained with anti-CD19 mAb (Phycoethrin 1D3; BD Biosciences), fixed in 4% formaldehyde, permeabilized in 0.1% saponin, and stained in permeabilization buffer with the following FITC-conjugated mAbs: anti–γ-interferon (IFN-γ) (XMG1.2; eBioscience), anti–interleukin (IL)-4 (BVD6-24G2; eBioscience), and anti–IL-10 (JES5-16E3; BD Pharmingen). Stained cells were then analyzed by flow cytometry.

T-cell proliferation assays.

Splenic and islet-infiltrating mononuclear cell suspensions were stained with anti-CD3 (FITC 17A2; BD Biosciences) and anti-CD19 (PE 1D3; BD Biosciences) and then sorted using the FACS Vantage cytometer (BD Biosciences). In all the assays, purity of T-cell–and B-cell–sorted populations was always >98%. Purified T-cells (5 × 106/ml) were labeled with an equal volume of 1.25 μmol/l carboxy-fluorescein diacetate succinimidyl ester (CFSE) (5-[and 6]-CFSE; Molecular Probes, Eugene, OR) for 10 min at room temperature and then quenched with the same volume with heat-inactivated fetal bovine serum for 10 min. Then T-cells were washed twice with PBS and resuspended in culture medium. T- and B-cells were cocultured (200 μl supplemented culture medium) in plate-bound 10 μg/ml anti-CD3 mAb (145-2C11; BD Pharmingen) or soluble 5 μg/ml anti-CD3 alone or supplemented with islet extracts obtained by freeze-thaw cycles (10 islets/well) and/or 10 μg/ml anti-CD40 mAb (3/23; BD Pharmingen). For a positive control of T-cell proliferation, purified T-cells were cultured in a plate-bound anti-CD3 mAb or cocultured with unfractionated islet-infiltrating APCs plus islet extract. Unfractionated islet-infiltrating APCs were obtained by selecting negative anti-CD3 mononuclear cell populations by cell sorting. After 3 days of culture, cells were harvested and analyzed by flow cytometry.

Statistical analyses.

All data are shown as mean ± SE unless otherwise indicated. Statistical analyses were performed using the Mann-Whitney U test (SPSS, Chicago, IL, and GraphPad Prism, Graphpad Software, San Diego, CA).

The main objective of the present study was to characterize the islet-infiltrating B-cell population in NOD and (NOD×NOR)F1 mice. Having used peripheral B-cells as reference cells, we previously characterized this population resident in the spleen. Splenic B-cells from 12-week female NOD, NOR, and BALB/c mice (used as controls) were stained with a combination of surface markers indicative of activation, maturation, and B-cell lineage and were subsequently analyzed by flow cytometry.

Splenic B-cells from NOD and NOR mice show phenotypic signals of autoimmune activation.

A significant increase in the IgMhighIgDlow/neg CD21+ B-cell population was detected in the spleen of NOD mice compared with Balb/c and NOR mice (Fig. 1A), thus confirming previous studies indicating that the marginal zone B-cell population is increased in the spleens of NOD mice (15,16).

No significant differences were found in the expression of CD21, CD40, CD45R, or CD86 molecules in splenic B-cells of the three mouse strains. However, flow cytometric analysis showed that splenic B-cells from NOD and NOR mice expressed higher levels of CD24, CD44, and major histocompatibility complex (MHC) class I molecules (H-2kd) than those from Balb/c mice (Fig. 1B and C). CD24 and CD44 levels were significantly higher in NOD than in NOR mice, suggesting a higher B-cell activation in NOD mice. Interestingly, the expression levels of the B-cell coreceptor molecule CD19 were 50% higher in NOD and NOR splenic B-cells than in those of Balb/c mice. Furthermore, the percentage of splenic B-cells CD5+ (CD11b) in NOD and NOR mice was higher than in Balb/c mice (24–30 vs. 7%). As these data suggest that splenic B-cells are activated similarly in NOD and NOR mice, this must reflect a consequence of their background and not a function of diabetes progression.

The percentage of islet-infiltrating B-cells increases with the progression of the disease influenced by sex traits.

Next we studied the fate of B-cells during type 1 diabetes development. We assessed the percentage of B-cells in the spleens of NOR and NOD mice at different stages of the disease (at weeks 5, 7, 9, and 12), in adult resistant mice (at 32 weeks), and in recently diabetic mice. The islet infiltration analysis was pursued in NOD and (NOD×NOR)F1 mice, since NOR mice only develop mild peri-insulitis.

The percentage of splenic B-cells remained quite constant during the progression of the disease in NOD mice, with values between 50 and 60% in most cases (Fig. 2A). Similar results were found in NOR mice. However, the percentage of islet-infiltrating B-cells tended to increase during this period (Fig. 2B), especially in NOD and (NOD×NOR)F1 female mice, and the highest values (almost 50%) were detected at 12 weeks. This may be crucial in the disease development, since it occurs just before the onset of diabetes. After that age, the percentage of islet-infiltrating B-cells decreases with age. To elucidate whether B-cells accumulate into the pancreatic islets as a consequence of their recruitment or their proliferation in situ, we double-stained B-cells to analyze the expression of the cell proliferation marker ki67. However, we did not find B-cell proliferation in the pancreatic islets (Fig. 3). Therefore, these data indicate that B-cell recruitment into pancreatic islets is strongly influenced by gender traits and suggest that this may be essential to determine the progression of insulitis but not disease development.

Phenotypic characterization of islet-infiltrating B-cells reveals a silenced population.

To further characterize B-cell infiltrate in islets, we carried out new surface marker analysis on the cell population of 12-week NOD and (NOD×NOR)F1 female mice. To analyze B-cells in their native state, we performed all the stainings as quickly as possible after islet isolation. We considered the possible effects of collagenase digestion used for islet isolation protocol on the surface antigens, which is why we overdigested splenic B-cells with collagenase and then analyzed each surface marker to evaluate their integrity. The results indicated that only CD21, CD44, and CD45R/B220 molecules were affected by collagenase digestion; we therefore did not use them in the phenotype study of islet-infiltrating B-cells (data not shown).

The immunoglobulin isotype analysis of islet-infiltrating B-cells indicated that ∼90% of these cells were IgM IgD double-positive B-cells (Fig. 4), whereas ∼3% were positive for IgG1. Only a few B-cells expressed other isotypes (e.g., IgG2, IgG3, and IgA), and their percentage could not to be accurately determined (≤1%) (data not shown).

Cytometric analysis of islet-infiltrating B-cells also showed a slight but significant increase in the percentage of CD69+ (11 vs. 5% in splenic B-cells) and CD86+ cells (7 vs. 2.5% in the spleen) (Fig. 5A), but no differences were found in the expression of CD80 molecules (both islet-infiltrating and splenic B-cells were CD80-negative [data not shown]). Moreover, the percentage of CD5+ B-cells was similar to that found in the spleen. Interestingly, we found that CD19 expression decreased by >60% in the whole infiltrating B-cell population compared with that from the spleen (Fig. 5B and C), showing a possible hyporesponsive status of these cells, given the relevance of CD19 in regulating B-cell activation. CD19 downmodulation was more pronounced during the progression of the disease (Fig. 6). The decrease in CD19 expression coexisted with a significant increase in MHC class I and II molecule expression (Fig. 5D). The population of islet-infiltrating B-cells also showed a decrease in CD24 surface molecules (Fig. 5D) and mIgD expression but not in mIgM expression (Fig. 4). No significant differences were found in the expression levels of CD40 between splenic and islet-infiltrating B-cells. Similar results were observed in the phenotype analysis of (NOD×NOR)F1 mice. In general, these results suggest that islet-infiltrating B-cells are not naive cells, as they had undergone upregulation of the antigen presentation machinery, possibly as a result of the encounter with their specific antigen. In that case, islet-infiltrating B-cells may present the captured autoantigen in the absence of costimulatory molecules, and therefore they may not be able to provide adequate stimulatory signals to activate T-cells. On the contrary, it is also possible that this B-cell phenotype may be secondary to the encounter with activated T-cells, i.e., by bystander help, or to the proinflammatory status present at the islet milieu. As a result, despite their antigenic specificity, the behavior of islet-infiltrating B-cells may be impaired, and thus these cells are unable to respond normally to antigen-specific stimulation.

Cytokine expression profile in islet-infiltrating B-cells is similar to that in their splenic counterparts.

As the secretion of certain cytokines by islet-infiltrating B-cells may act in situ on other mononuclear cells, we analyzed the possible differential expression of IFN-γ, IL-10, and IL-4 cytokines. Cytokine staining showed that the percentage of B-cells producing each type of cytokine was similar in both the islet infiltrate and spleen of NOD female mice (3% IFN-γ+, 1% IL-10+, and almost undetectable for IL-4+) (Fig. 7). Similar results were found in 12-week female (NOD×NOR)F1 mice (data not shown). Thus, no differences were found in the cytokine profile of islet-infiltrating B-cells in either NOD or (NOD×NOR)F1 mice.

Islet-infiltrating B-cells develop an important costimulator phenotype following in vitro stimulation and are thus antigen-experienced B-cells.

Although the phenotype expressed by islet-infiltrating B-cells does not correspond to that classically defined for anergic B-cells (Fig. 5), in view of CD19 downmodulation we considered it important to explore this hypothesis. Therefore, islet-infiltrating B-cells from 12-week NOD female mice were cultured with different stimulating agents. LPS was used to study the response to T-cell–independent polyclonal stimulation, whereas anti-CD40 plus IL-4 were used to simulate a T-cell–dependent response. Islet-infiltrating B-cells did not show proliferation arrest, which is typical of anergic cells, but reached higher levels of antigen-presenting MHC class I and II molecules, CD86 (but not CD80 [data not shown]), and activation markers (e.g., CD69 and CD44) than splenic B-cells (Fig. 8). This difference was more obvious when cultured in anti-CD40 plus IL-4 (or anti-CD40 alone [data not shown]). Interestingly, the expression of CD19 in islet-infiltrating B-cells cultured in LPS or anti-CD40 plus IL-4 was always lower than in splenic B-cells. This different reaction of islet-infiltrating B-cells to in vitro stimulating agents clearly indicates that they are antigen experienced. Note that changes in the expression levels of some surface molecules (i.e., CD19, H-2Kd, CD86) were detected even when islet-infiltrating B-cells were cultured in complete culture medium alone out of the islet milieu, thus supporting the idea that they are somehow silenced in the infiltrate environment. However, this silenced status is easily reversible, becoming potentially antigen-presenting cells, particularly under T-cell–dependent stimulus.

Islet-infiltrating B-cells only induce T-cell proliferation in vitro in the presence of anti-CD3.

To establish whether islet-infiltrating B-cells could act as APCs for islet-infiltrating T-cells, purified T- and B-cells (>98% purity) from spleen or islet infiltration were cocultured under different stimulating conditions, after which they were harvested and analyzed by cytometry. T-cell proliferation was analyzed by CFSE cytometric assay because it allowed us to study the proliferation of small cell populations. The results showed that islet-infiltrating B-cells cannot induce polyclonal proliferation of T-cells by themselves (Fig. 9), even when anti-CD40 mAb or islet extract is added to the culture. Hence, B-cells cannot present the specific antigen or provide a correct costimulatory signal to T-cells. We conducted further experiments culturing T- and B-cells in plate-bound anti-CD3 mAb, soluble anti-CD3, or soluble anti-CD3 plus anti-CD40 or islet extracts. The results indicated that neither splenic nor islet-infiltrating B-cells inhibit plate-bound anti-CD3–induced T-cell proliferation; however, both B-cell subsets act as accessory cells to promote similar splenic and islet-infiltrating T-cell proliferation in the presence of soluble anti-CD3. No significant differences were found when we conducted these same proliferative studies adding anti-CD40 mAb or islet extract to the culture. Similar results were obtained when we analyzed (NOD×NOR)F1 female mice (data not shown). Therefore, under in vitro conditions, islet-infiltrating B-cells from both NOD and (NOD×NOR)F1 mice cannot restrain T-cell proliferation. On the contrary, they can only provide adequate signals to induce polyclonal T-cell proliferation in the presence of the mitogenic soluble anti-CD3 mAb.

During the last decade, many studies in NOD mice demonstrated that B-cells are essential in type 1 diabetes progression (3). How, when, and where they provide this diabetogenic activity remains unclear. B-cells may induce diabetogenesis by presenting autoantigens to β-cell CTLs and/or by producing pathogenic autoantibodies in the spleen, regional lymph nodes, and/or inside pancreatic islets at any stage of the disease. The aim of this study was to phenotypically and functionally characterize the B-cell population in spleen and islet infiltrate to determine its possible role in the disease.

Our studies revealed that the splenic marginal-zone B-cell population is increased in the spleens of NOD but not NOR mice. These results support recent studies by Rolf et al. (16) indicating that this trait maps to diabetes susceptibility Idd9 and Idd11 loci in NOD mice (which are not present in NOR mice) and that the increase in the marginal-zone B-cell population is related to type 1 diabetes development.

Signs of B-cell activation have been observed in the spleen of 1-month-old NOD mice (17). Some authors have described that splenic B-cells highly express CD19, CD21, CD40, and MCH class II molecules, which may suggest a role in the activation of autoreactive T-cells (18). In our studies, we confirmed splenic B-cell activation by an increase in cell surface expression of CD19, CD44, CD24, and H-2Kd molecules in this cell population in both NOD and NOR compared with Balb/c mice. Moreover, the percentage of splenic CD5+ B-cells in NOD and NOR mice was clearly higher than in Balb/c mice. Since they are Mac-1 negative, this CD5+ B-cell population corresponds to B-2 cells, in which, most likely, CD5 has been selectively induced to provide inhibitory signals after recognition of autoantigen (19,20). These so-called “induced CD5+ B-cells” are mainly found in the mantle zone of germinal centers (21). Although CD5+ B-cells have been related to autoimmunity in humans (22) and in mice (23), their role in autoimmune diseases has not been demonstrated. Interestingly, these CD5+ B-cells are found both in NOD and NOR mice, thus indicating that their presence is a consequence of the autoreactive background of both mouse strains and not of diabetes progression.

It has been shown that systemic (24) or peritoneal-selective (25) depletion of the B-cell population in wild-type NOD mice, or their absence in NOD.Igμnull mice (2628), results in type 1 diabetes and/or insulitis resistance. Hence, one of the most interesting findings of our study shows that during the pre-diabetic period, B-cells accumulate into the islets influenced by sex traits in both NOD and (NOD×NOR)F1 mice. The fact that the incidence of diabetes is higher in NOD and (NOD×NOR)F1 female mice than male strongly suggests that B-cell recruitment to the islets is one of the key features, though not critical, in the progression of the disease to overt diabetes.

Compared with splenic, the phenotype of most islet-infiltrating B-cells is characterized by downmodulation of CD19, a low expression of IgD (but not IgM), low levels of CD24, an upregulation of MHC class I and II molecules, and similar levels of CD40. CD19 is a component of the stimulatory coreceptor of B-cells that enhances B-cell receptor signal and is essential for the response of mature B-cells to antigenic challenge (particularly to thymus-dependent antigens). Therefore, the formation of germinal centers, the production of high-affinity antibodies, and the development of memory B-cells are impaired in CD19-deficient mice (29). On the contrary, CD19 overexpression induces autoantibody production in systemic sclerosis patients (30) and in CD19 transgenic mice (31) not genetically predisposed to autoimmunity, demonstrating the role of this molecule in the mechanisms of tolerance. Moreover, signaling pathways via CD19 may affect antigen processing and may enhance or inhibit the endocytic pathways, depending on the signal intensity (32). Apparently, our results suggest that most islet-infiltrating B-cells are downregulated and cannot act as regular APCs and activate islet-infiltrating T-cells.

B-cells that recognize autoantigens are anergyzed as a mechanism of immune tolerance. The anergic B-cell phenotype downmodulates IgM, CD21, CD20, CD22, CD23, CD40, and l-selectin molecules and upregulates CD24, CD44, and MHC class II molecules (33). Though we did not analyze all of these markers (some of which were affected by collagenase digestion), this does not seem to be the case of islet-infiltrating B-cells, which showed lower CD24 levels and similar IgM and CD40 levels compared with splenic B-cells. Moreover, they developed a strong APC phenotype after being cultured in LPS or anti-CD40. Note that, following the in vitro stimulation, the upregulation of MHC, CD44, CD69, and CD86 molecules in islet-infiltrating B-cells was even higher than in splenic B-cells, strongly suggesting that they are antigen experienced. In fact, several authors found that islet-infiltrating B-cells were significantly more responsive to anti-IgMF(ab′)2 or IL-4 stimulations than splenic B-cells in NOD mice (34). Why, then, do islet-infiltrating B-cells downmodulate CD19 molecules? The possible effect of collagenase on CD19 was ruled out by digesting splenic B-cells with this enzyme for twice the period of time than is usual in islet isolation. Interestingly, after islet isolation, islet-infiltrating B-cells that leave the islets increase CD19 levels even when there is no stimulation, thus strongly suggesting that soluble factors, such as cytokines, released in the islet environment are responsible for the islet-infiltrating B-cell phenotype. However, after being challenged with LPS or anti-CD40, islet-infiltrating B-cells still show lower levels of CD19 than splenic B-cells. Physiological downmodulation of CD19 has been described during plasmatic cell development (35), but apparently this does not occur in islet-infiltrating B-cells. It has been described that the CD19 molecule is downmodulated by low-affinity FcγRII (CD32) stimulation (36). In addition, FcγRII acts as an inhibitory receptor in B-cells, thus preventing their activation. Therefore, this mechanism may also be involved in the downmodulation of CD19 in islet-infiltrating B-cells.

It has been shown that a significant number of B-1 cells accumulate in the islet infiltrate and play a critical role in the development of insulitis and diabetes (25). However, we found that most islet-infiltrating CD5+ B-cells were CD11b negative, thus indicating that they were not B-1 cells. Moreover, there were no differences in the percentage of CD5+ B-cells between the islet infiltrate and the spleen. Our results are consistent with a recently published report (38) suggesting that the B-1 cell population is not present in the islet infiltrate.

Only a few islet-infiltrating B-cells showed signs of activation (11% were CD69+ and 7% CD86+). These results do not agree with those of Hussain et al. (34), wherein 80% of islet-infiltrating B-cells were CD69+ and CD86+. We performed all staining procedures immediately after islet isolation, whereas in the study by Hussain et al., phenotypic analysis was carried out following overnight culture of islet infiltrates. We have already described a significant increase in CD19, CD44, CD69, CD86, and MHC class I molecules after a short-period culture of islet-infiltrating B-cells. Most likely, the observed differences are caused by the different methodologies used.

Islet-infiltrating B-cells have been assigned the ability to activate islet-infiltrating T-cells (37). Our findings indicate the opposite. In our studies, islet-infiltrating B-cells did not induce the proliferation of their counterpart T-cells by themselves, even when they were activated with anti-CD40, or in the presence of islet extract. However, this B-cell population fully induced T-cell activation when acting as accessory cells. These results strongly suggest that islet-infiltrating B-cells do not present the appropriate antigens to islet-infiltrating T-cells. In this regard, we have recently reported that only a few islet-infiltrating B-cells may be islet-antigen restricted (38), supporting the idea that islet-infiltrating B-cells do not act as APCs in situ.

In conclusion, islet-infiltrating B-cells accumulate into the islets influenced by sex traits in NOD and (NOD × NOR)F1 mice. Phenotype and functional characteristics of islet-infiltrating B-cells suggest that in the proinflammatory islet milieu there are also immune regulatory mediators that may prevent their activation. Our results strongly suggest that B-cells do not induce islet-infiltrating T-cell activation in situ but may have other unknown roles in the development of the disease.

FIG. 1.

Phenotypic characterization of splenic B-cells in 12-week-old females; differences among NOD, NOR, and BALB/c mouse strains. Splenic cells were stained for surface markers and analyzed by flow cytometry. CD19 was used as a B-cell population marker. A: IgMlowIgDhigh (mature B-cells), IgMhighIgDlow/neg (marginal zone and transitional T1 B-cells), and IgMhighIgDhigh (transitional T2 B-cells) percentages. B: B-cells with positive staining for CD86 or CD5 percentages. C: Surface expression levels of CD19, CD45R/B220, CD40, CD21, CD24, CD44, and MHC class I molecules. Data correspond to average values from 3–6 mice per group. Bars show the SE of the mean values. *Statistically significant differences according to Mann-Whitney U test. P < 0.05. MFI, median fluorescence intensity.

FIG. 1.

Phenotypic characterization of splenic B-cells in 12-week-old females; differences among NOD, NOR, and BALB/c mouse strains. Splenic cells were stained for surface markers and analyzed by flow cytometry. CD19 was used as a B-cell population marker. A: IgMlowIgDhigh (mature B-cells), IgMhighIgDlow/neg (marginal zone and transitional T1 B-cells), and IgMhighIgDhigh (transitional T2 B-cells) percentages. B: B-cells with positive staining for CD86 or CD5 percentages. C: Surface expression levels of CD19, CD45R/B220, CD40, CD21, CD24, CD44, and MHC class I molecules. Data correspond to average values from 3–6 mice per group. Bars show the SE of the mean values. *Statistically significant differences according to Mann-Whitney U test. P < 0.05. MFI, median fluorescence intensity.

Close modal
FIG. 2.

Development of B-cell populations with age. A: Splenocytes from NOD and NOR mice were stained with anti-CD19 and analyzed by flow cytometry to measure the percentage of B-cells within lymphocyte population. In all the groups analyzed, the percentage of splenic B-cells remained constant for life. B: Islet infiltrates from NOD and (NOD×NOR)F1 male and female mice were analyzed as previously described. There are no data from 5-week-old (NOD×NOR)F1 mice, since not enough infiltrating cells were obtained. The percentage of islet-infiltrating B-cells increased with age. Compared with that in males, the percentage of B-cells significantly increased at 12 weeks in NOD and (NOD × NOR)F1 female mice (P < 0.015). Note that the same percentage (∼50%) of B-cells was seen in NOD and (NOD×NOR)F1 female mice at 12 weeks of age, despite their diabetes susceptibility/resistance. Data correspond to average values from 3–6 mice per group. Bars show the SE of the means. *Statistically significant difference.

FIG. 2.

Development of B-cell populations with age. A: Splenocytes from NOD and NOR mice were stained with anti-CD19 and analyzed by flow cytometry to measure the percentage of B-cells within lymphocyte population. In all the groups analyzed, the percentage of splenic B-cells remained constant for life. B: Islet infiltrates from NOD and (NOD×NOR)F1 male and female mice were analyzed as previously described. There are no data from 5-week-old (NOD×NOR)F1 mice, since not enough infiltrating cells were obtained. The percentage of islet-infiltrating B-cells increased with age. Compared with that in males, the percentage of B-cells significantly increased at 12 weeks in NOD and (NOD × NOR)F1 female mice (P < 0.015). Note that the same percentage (∼50%) of B-cells was seen in NOD and (NOD×NOR)F1 female mice at 12 weeks of age, despite their diabetes susceptibility/resistance. Data correspond to average values from 3–6 mice per group. Bars show the SE of the means. *Statistically significant difference.

Close modal
FIG. 3.

Islet-infiltrating B-cells do not proliferate in situ. Double immunofluorescence staining of B-cells with anti-CD45R/B220 mAb (green) and anti-ki67 (red) on cryosections from pancreas of a 12-week-old NOD female mouse. In the figure, an islet with a moderate infiltration is shown. Lack of coincidence of both stainings on the same cells indicates that islet-infiltrating B-cells do not proliferate in situ.

FIG. 3.

Islet-infiltrating B-cells do not proliferate in situ. Double immunofluorescence staining of B-cells with anti-CD45R/B220 mAb (green) and anti-ki67 (red) on cryosections from pancreas of a 12-week-old NOD female mouse. In the figure, an islet with a moderate infiltration is shown. Lack of coincidence of both stainings on the same cells indicates that islet-infiltrating B-cells do not proliferate in situ.

Close modal
FIG. 4.

IgD and IgM profiles of splenic and islet-infiltrating B-cells. Representative data from a 12-week-old female NOD mouse.

FIG. 4.

IgD and IgM profiles of splenic and islet-infiltrating B-cells. Representative data from a 12-week-old female NOD mouse.

Close modal
FIG. 5.

Phenotypic characterization of splenic and islet-infiltrating B-cells in 12-week-old NOD female mice. A: Percentage of CD86+, CD69+, or CD5+ B-cells in the spleen and in the islet infiltrate. B and C: Median fluorescence intensity (MFI) for CD19, CD40, CD24, and MHC molecules in splenic and islet-infiltrating B-cells. Dotted line in C indicates the median fluorescence intensity of negative controls. Data correspond to average values from a minimum of six mice group. Bars show the SE of the means. *Statistically significant differences between data obtained from splenic and islet-infiltrating B-cells according to Mann-Whitney U test. P < 0.02. D: CD19, CD24, H-2Ag7, and H-2Kd profiles of B-cells from islet infiltrate (thick black line) and spleen (digested [gray line] and nondigested [thin black line] with collagenase).

FIG. 5.

Phenotypic characterization of splenic and islet-infiltrating B-cells in 12-week-old NOD female mice. A: Percentage of CD86+, CD69+, or CD5+ B-cells in the spleen and in the islet infiltrate. B and C: Median fluorescence intensity (MFI) for CD19, CD40, CD24, and MHC molecules in splenic and islet-infiltrating B-cells. Dotted line in C indicates the median fluorescence intensity of negative controls. Data correspond to average values from a minimum of six mice group. Bars show the SE of the means. *Statistically significant differences between data obtained from splenic and islet-infiltrating B-cells according to Mann-Whitney U test. P < 0.02. D: CD19, CD24, H-2Ag7, and H-2Kd profiles of B-cells from islet infiltrate (thick black line) and spleen (digested [gray line] and nondigested [thin black line] with collagenase).

Close modal
FIG. 6.

Significant downregulation of CD19 in islet-infiltrating B-cells during type 1 diabetes (T1D) development. Median fluorescence intensity (MFI) for CD19 in splenic (of NOD and NOR mice) and islet-infiltrating (of NOD and [NOD×NOR]F1 mice) B-cells at different stages of disease progression. Data correspond to average values from three mice per group. Bars show the SE of the means.

FIG. 6.

Significant downregulation of CD19 in islet-infiltrating B-cells during type 1 diabetes (T1D) development. Median fluorescence intensity (MFI) for CD19 in splenic (of NOD and NOR mice) and islet-infiltrating (of NOD and [NOD×NOR]F1 mice) B-cells at different stages of disease progression. Data correspond to average values from three mice per group. Bars show the SE of the means.

Close modal
FIG. 7.

Cytokine production profile in splenic and islet-infiltrating B-cells from NOD mice. Splenic and islet-infiltrate mononuclear cells were stimulated with phorbol myristic acid plus ionomycin for 5 h; meanwhile, cytokine secretion was blocked with monensin. After staining with anti-CD19, cells were fixed, permeabilized, and intracellularly stained with anti–IFN-γ, anti–IL-4, or anti–IL-10 cytokine mAbs. Control antibodies showing the same isotype were used to adjust positive threshold. Data correspond to representative values from 3–6 mice/group. The percentage of B-cells (CD19+) positive for every cytokine is indicated.

FIG. 7.

Cytokine production profile in splenic and islet-infiltrating B-cells from NOD mice. Splenic and islet-infiltrate mononuclear cells were stimulated with phorbol myristic acid plus ionomycin for 5 h; meanwhile, cytokine secretion was blocked with monensin. After staining with anti-CD19, cells were fixed, permeabilized, and intracellularly stained with anti–IFN-γ, anti–IL-4, or anti–IL-10 cytokine mAbs. Control antibodies showing the same isotype were used to adjust positive threshold. Data correspond to representative values from 3–6 mice/group. The percentage of B-cells (CD19+) positive for every cytokine is indicated.

Close modal
FIG. 8.

Islet-infiltrating B-cells develop a pronounced costimulator phenotype after in vitro stimulation. Splenocytes and pancreatic islets were cultured in complete culture medium alone or supplemented with either LPS or anti-CD40 and IL-4 for 2 days. At times 0, 24, and 48 h, cultured cells were harvested and their surface expression levels of CD19, CD44, CD69, MHC, and costimulatory molecules analyzed by flow cytometry. Data corresponds to median fluorescence intensity levels on cultured B-cells of at least three independent assays. Each assay was done in triplicate.

FIG. 8.

Islet-infiltrating B-cells develop a pronounced costimulator phenotype after in vitro stimulation. Splenocytes and pancreatic islets were cultured in complete culture medium alone or supplemented with either LPS or anti-CD40 and IL-4 for 2 days. At times 0, 24, and 48 h, cultured cells were harvested and their surface expression levels of CD19, CD44, CD69, MHC, and costimulatory molecules analyzed by flow cytometry. Data corresponds to median fluorescence intensity levels on cultured B-cells of at least three independent assays. Each assay was done in triplicate.

Close modal
FIG. 9.

T-cell response to accessory functions of splenic and islet-infiltrating B-cells. The effect of B-cells on T-cell proliferation was assessed by culturing 4 × 104 splenic or islet-infiltrating CD19+ cells with 4 × 104 splenic or islet-infiltrating CD3+ cells for 72 h. Cultures were supplemented with either plate-bound anti-CD3 to assess the inhibitory ability of B-cells or soluble anti-CD3 to measure the ability of these cells to act as accessory cells. Costimulation of B-cells with anti-CD40 and islet extract was also analyzed. Columns represent the mean of the percentage of proliferating T-cells of at least two independent assays. Bars show the SE of the mean values. Solid and dotted lines indicate the percentage of proliferating T-cells in cultures with plate-bound anti-CD3 (without B-cells) or with unfractionated islet-infiltrating APCs plus islet extract, respectively.

FIG. 9.

T-cell response to accessory functions of splenic and islet-infiltrating B-cells. The effect of B-cells on T-cell proliferation was assessed by culturing 4 × 104 splenic or islet-infiltrating CD19+ cells with 4 × 104 splenic or islet-infiltrating CD3+ cells for 72 h. Cultures were supplemented with either plate-bound anti-CD3 to assess the inhibitory ability of B-cells or soluble anti-CD3 to measure the ability of these cells to act as accessory cells. Costimulation of B-cells with anti-CD40 and islet extract was also analyzed. Columns represent the mean of the percentage of proliferating T-cells of at least two independent assays. Bars show the SE of the mean values. Solid and dotted lines indicate the percentage of proliferating T-cells in cultures with plate-bound anti-CD3 (without B-cells) or with unfractionated islet-infiltrating APCs plus islet extract, respectively.

Close modal

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

This work was supported by a grant from the Fondo de Investigaciones Sanitarias of the Spanish National Institute of Health (FIS 00/0447 and FIS 01/1128). M.C.P. was supported by a predoctoral CIRIT fellowship (2001FI 0002) from the Catalan Government. A.A. was supported by a BEFI predoctoral fellowship (01/9065) from the Instituto Carlos III of Spanish National Institute of Health. R.M.A. and X.P. were supported by a Juvenile Diabetes Research Foundation research grant (1-2002-724). M.V.-P. is a researcher of the Fondo de Investigaciones Sanitarias of Spanish National Institute of Health. J.V. is an associate professor of the Serra-Hunter Programme from the Catalan Government.

We thank M.A. Fernandez for technical assistance and Miss D. Cullell-Young for editorial assistance.

1.
Tisch R, McDevitt HO: Insulin-dependent diabetes mellitus.
Cell
85
:
291
–297,
1996
2.
Kikutani H, Makino S: The murine autoimmune diabetes model: NOD and related strains.
Adv Immunol
51
:
285
–322,
1992
3.
Wong FS, Wen L: B cells in autoimmune diabetes.
Rev Diabet Stud
2
:
121
–135,
2005
4.
Lieberman SM, DiLorenzo TP: A comprehensive guide to antibody and T-cell responses in type 1 diabetes.
Tissue Antigens
62
:
359
–377,
2003
5.
Pietropaolo M, Eisenbarth GS: Autoantibodies in human diabetes.
Curr Dir Autoimmun
4
:
252
–282,
2001
6.
Bonifacio E, Atkinson M, Eisenbarth G, Serreze D, Kay TWH, Lee-Chan E, Singh B: International workshop on lessons from animal models for human type 1 diabetes: identification of insulin but not glutamic acid decarboxylase or IA-2 as specific autoantigens of humoral autoimmunity in nonobese diabetic mice.
Diabetes
50
:
2451
–2458,
2001
7.
Greeley SA, Katsumata M, Yu L, Eisenbarth GS, Moore DJ, Goodarzi H, Barker CF, Naji A, Noorchashm H: Elimination of maternally transmitted autoantibodies prevents diabetes in nonobese diabetic mice.
Nat Med
8
:
399
–402,
2002
8.
Facone M, Lee J, Patstone G, Yeung B, Sarvetnick N: B-lymphocytes are crucial antigen-presenting cells in the pathogenic autoimmune response to GAD65 antigen in nonobese diabetic mice.
J Immunol
161
:
1163
–1168,
1998
9.
Serreze DV, Fleming SA, Chapman HD, Richard SD, Leiter EH, Tisch RM: B lymphocytes are critical antigen-presenting cells for the initiation of T cell-mediated autoimmune diabetes in nonobese diabetic mice.
J Immunol
161
:
3912
–3918,
1998
10.
Noorchashm H, Lieu YK, Noorchashm N, Rostami SY, Greeley SAS, Schlachterman A, Song HK, Noto LE, Jevnikar AM, Barker CF, Naji A: I-Ag7-mediated antigen presentation by B lymphocytes is critical in overcoming a checkpoint in T cell tolerance to islet beta cells of nonobese diabetic mice.
J Immunol
163
:
743
–750,
1999
11.
Tian J, Zekzer D, Hanssen L, Lu Y, Olcott A, Kaufman DL: Lipopolysaccharide-activated B cells down-regulate Th1 immunity and prevent autoimmune diabetes in nonobese diabetic mice.
J Immunol
167
:
1081
–1089,
2001
12.
Serreze DV, Prochazka M, Reifsnyder PC, Bridgett MM, Leiter EH: Use of recombinant congenic and congenic strains of NOD mice to identify a new insulin-dependent diabetes resistance gene.
J Exp Med
180
:
1553
–1558,
1994
13.
Prochazka M, Serreze DV, Frankel WN, Leiter EH: NOR/Lt mice: MHC-matched diabetes-resistant control strain for NOD mice.
Diabetes
41
:
98
–106,
1992
14.
Verdaguer J, Yoon JW, Anderson B, Averill N, Utsugi T, Park BJ, Santamaria P: Acceleration of spontaneous diabetes in TCR-beta-transgenic nonobese diabetic mice by beta-cell cytotoxic CD8+ T cells expressing identical endogenous TCR-alpha chains.
J Immunol
157
:
4726
–4735,
1996
15.
Noorchashm H, Moore DJ, Lieu YK, Noorchashm N, Schlachterman A, Song HK, Barker CF, Naji A: Contribution of the innate immune system to autoimmune diabetes: a role for the CR1/CR2 complement receptors.
Cell Immunol
195
:
75
–79,
1999
16.
Rolf J, Motta V, Duarte N, Lundholm M, Berntman E, Bergman M-L, Sorokin L, Cardell SL, Berntman E: The enlarged population of MZ/CD1dhigh B lymphocytes in nonobese diabetic mice maps to diabetes susceptibility region Idd11.
J Immunol
174
:
4821
–4827,
2005
17.
Luzina IG, Atamas SP, Storrer CE, daSilva LC, Kelsoe G, Papadimitriou JC, Handwerger BS: Spontaneous formation of germinal centers in autoimmune mice.
J Leukoc Biol
70
:
578
–584,
2001
18.
Moore DJ, Noorchashm H, Lin TH, Greeley SA, Naji A: NOD B-cells are insufficient to incite T-cell–mediated anti-islet autoimmunity.
Diabetes
54
:
2019
–2025,
2005
19.
Pers JO, Jamin C, Predine-Hug F, Lydyard P, Youinou P: The role of CD5-expressing B cells in health and disease.
Int J Mol Med
3
:
239
–245,
1999
20.
Berland R, Wortis HH: Origins and functions of B-1 cells with notes on the role of CD5.
Annu Rev Immunol
20
:
253
–300,
2002
21.
Youinou P, Jamin C, Lydyard PM: CD5 expression in human B-cell populations.
Immunol Today
20
:
312
–316,
1999
22.
Talal N, Dauphinee M, Ahmed SA: CD5 B cells in autoimmunity.
Ann N Y Acad Sci
651
:
551
–556,
1992
23.
Hayakawa K, Hardy RR: Development and function of B-1 cells.
Curr Opin Immunol
12
:
346
–53,
2000
24.
Noorchashm H, Noorchashm N, Kern J, Rostami SY, Barker CF, Naji A: B-cells are required for initiation of insulitis and sialitis in nonobese diabetic mice.
Diabetes
46
:
941
–946,
1997
25.
Kendall PL, Woodward EJ, Hulbert C, Thomas JW: Peritoneal B cells govern the outcome of diabetes in non-obese diabetic mice.
Eur J Immunol
34
:
2387
–2395,
2004
26.
Serreze DV, Chapman HD, Varnum DS, Hanson MS, Reifsnyder PC, Richard SD, Fleming SA, Leiter EH, Shultz LD: B lymphocytes are essential for the initiation of T cell-mediated autoimmune diabetes: analysis of a new “speed congenic” stock of NOD.Igμnull mice.
J Exp Med
184
:
2049
–2053,
1996
27.
Yang M, Charlton B, Gautam AM: Development of insulitis and diabetes in B cell-deficient NOD mice.
J Autoimmun
10
:
257
–260,
1997
28.
Akashi T, Nagafuchi S, Anzai K, Kondo S, Kitamura D, Wakana S, Ono J, Kikuchi M, Niho Y, Watanabe T: Direct evidence for the contribution of B cells to the progression of insulitis and the development of diabetes in non-obese diabetic mice.
Int Immunol
9
:
1159
–1164,
1997
29.
Wang Y, Carter RH: CD19 regulates B cell maturation, proliferation, and positive selection in the FDC zone of murine splenic germinal centers.
Immunity
22
:
749
–761,
2005
30.
Sato S, Fujimoto M, Hasegawa M, Takehara K, Tedder TF: Altered B lymphocyte function induces systemic autoimmunity in systemic sclerosis.
Mol Immunol
41
:
1123
–1133,
2004
31.
Stohl W, Xu D, Kim KS, Tedder TF, Sato S: Humoral autoimmunity in mice overexpressing B cell surface CD19: vital role for MHC class II.
Clin Immunol
116
:
257
–264,
2005
32.
Wagle NM, Cheng P, Kim J, Sproul TW, Kausch KD, Pierce SK: B-lymphocyte signaling receptors and the control of class-II antigen processing.
Curr Top Microbiol Immunol
245
:
101
–126,
2000
33.
Noorchashm H, Bui A, Li HL, Eaton A, Mandik-Nayak L, Sokol C, Potts KM, Pure E, Erikson J: Characterization of anergic anti-DNA B cells: B cell anergy is a T cell-independent and potentially reversible process.
Int Immunol
11
:
765
–776,
1999
34.
Hussain S, Salojin KV, Delovitch TL: Hyperresponsiveness, resistance to B-cell receptor–dependent activation-induced cell death, and accumulation of hyperactivated B-cells in islets is associated with the onset of insulitis but not type 1 diabetes.
Diabetes
53
:
2003
–2011,
2004
35.
Calame KL, Lin KI, Tunyaplin C: Regulatory mechanisms that determine the development and function of plasma cells.
Annu Rev Immunol
21
:
205
–230,
2003
36.
Ikemoto T, Nakagawa T, Hatanaka M, Hasegawa M, Kageyama T, Hirano M, Shimizu A: My4+/LeuM3- molecule and CD19 antigen are down-modulate by low affinity Fc gamma receptor II (CD32) stimulation on CD56-positive B-lymphoma cells.
Leuk Lymphoma
39
:
157
–164,
2000
37.
Hussain S, Delovitch TL: Dysregulated B7-1 and B7-2 expression on nonobese diabetic mouse B cells is associated with increased T cell costimulation and the development of insulitis.
J Immunol
174
:
680
–687,
2005
38.
Carrillo J, Puertas MC, Alba A, Ampudia RM, Pastor X, Planas R, Riutort N, Alonso N, Pujol-Borrell R, Santamaria P, Vives-Pi M, Verdaguer J: Islet-infiltrating B-cells in nonobese diabetic mice predominantly target nervous system elements.
Diabetes
54
:
69
–77,
2005