Dendritic cells (DC) present Ag to naive T cells and are therefore pivotal in shaping immune responses. DC may either immunize or tolerize T cells. Humans with pancreatic islet autoimmunity at high risk for insulin-dependent diabetes mellitus (IDDM) present the opportunity to investigate DC in autoimmune disease. We compared DC phenotype and function in 12 euglycemic, asymptomatic IDDM relatives with islet autoimmunity and controls matched for age, sex, and MHC class II alleles. DC were generated from adherent peripheral blood cells by culture with granulocyte/macrophage-CSF and IL-4. The yield of DC was significantly lower in IDDM relatives than in controls. While the DC phenotype, HLA-DR+CD14, was expressed by ≥90% of the cells generated from relatives and controls, the proportion of cells that expressed CD1a and the costimulator molecules CD80 (B7-1) and CD86 (B7-2) was significantly lower in IDDM relatives. In addition, B7-1 and B7-2 expression per cell was significantly lower in IDDM relatives. These phenotypic changes were accompanied by reduced stimulation of autologous CD4 cells by DC from IDDM relatives. Similar findings were obtained in three recently diagnosed IDDM patients. These findings indicate that impairment of DC phenotype and function is a marker of islet autoimmunity and are consistent with a role for impaired DC function in the pathogenesis of autoimmune disease.

Destruction of insulin-producing pancreatic islet β cells in insulin-dependent diabetes mellitus (IDDM)3 is mediated by autoreactive T cells on a background of susceptibility MHC class II molecules, particularly HLA-DR3,4 (DQ2,8 in Caucasoids) (1). While islet Ag-specific T cells are present in individuals who develop IDDM, they can also be detected in healthy HLA-matched controls (2, 3, 4, 5). This implies that autoreactive T cells not deleted during development in the thymus are indifferent to peripheral autoantigens or are controlled by regulatory mechanisms. T cells are primed by dendritic cells (DC), the Ag-presenting capacity of which is reflected by high expression levels of MHC and costimulator CD80 (B7-1) and CD86 (B7-2) molecules. During development, presentation of self-peptides by DC in the thymus is responsible for the deletion by apoptosis of high affinity, autoreactive T cells (6). In addition to this central process of negative selection, DC in the periphery can either immunize or tolerize T cells depending on their expressed levels of Ag, costimulator, and other surface molecules (7, 8, 9) and their cytokine-hormonal milieu (10, 11, 12). It is reasonable, therefore, to assume that DC play an important role in the genesis of autoimmune disease.

In established clinical IDDM, Jansen et al. (13) found that monocyte-derived DC generated with triiodothyronine had a reduced ability to cluster and to elicit the autologous and allogeneic MLR. However, the phenotype of these DC was not defined and matching of control subjects for MHC molecules, necessary in evaluating APC function, was lacking. The phenotype and function of DC in humans at risk for autoimmune disease, including those with islet autoimmunity uncomplicated by insulin deficiency and metabolic dysfunction, are unknown. Investigation of DC function in humans has been facilitated by the ability to generate monocyte- or adherent cell-derived DC from peripheral blood by culture in the presence of granulocyte/macrophage-CSF (GM-CSF) and IL-4 (14, 15, 16) or IL-7 (17). We have analyzed the phenotype and function of peripheral blood monocyte-derived DC generated in GM-CSF/IL-4 from euglycemic, asymptomatic, first-degree IDDM relatives with underlying islet autoimmunity compared with age-, sex-, and MHC class II-matched controls.

First-degree relatives of IDDM patients (five males, seven females) positive for circulating Abs to islet Ags (Table I) were sex-matched, closely age-matched, and completely HLA class II-matched with healthy volunteer controls from the Australian Bone Marrow Donor Registry who were negative for Abs to islet Ags. The mean ages of at-risk IDDM relatives (25.1 yr; range, 8–50) and the control subjects (34.0 yr; range, 20–47) were not significantly different (p = 0.1). Subsequently, three patients with newly diagnosed (<3 mo) IDDM on insulin treatment and with well-controlled blood glucose levels were sex-, age-, and HLA-matched for study with three healthy controls (Table II). Subjects were bled from a forearm vein between 8:30 and 9:30 a.m. The study was approved by the Institution Human Ethics Committee.

Table I.

Characteristics of first-degree relatives at risk for IDDMa

SubjectAgeSexMHC Class II HLA DR;DQ AllelesRelationship to IDDM RelativeICA (<20 JDF U)IAA (<35 nU/ml)GAD Ab (<5 U)IA-2 Ab (<4 U)FPIR (mU/L)
11 3,4;2,8 Sibling 63 82 75 15 93 
48 3,4;2,7 Mother 160 92 89 −14 117 
44 3,12;2,7 Mother 160 −15 −1 260 
4,8;4,8 Sibling 20 760 49 100 62 
15 4,4;8,8 Sibling 75 120 42 88 234 
16 4,7;2,8 Sibling 25 3800 40 23 152 
15 4,4;8,8 Sibling 750 400 29 20 121 
16 3,3;2,2 Sibling 160 30 88 41 
46 4,4;8,8 Mother 129 90 49 89 44 
10 50 3,4;2,7 Mother 43 100 14 11 28 
11 14 2,4;1,8 Sibling 16 200 83 −5 151 
12 18 4,4;7,8 Sibling 50 44 86 −7 36 
SubjectAgeSexMHC Class II HLA DR;DQ AllelesRelationship to IDDM RelativeICA (<20 JDF U)IAA (<35 nU/ml)GAD Ab (<5 U)IA-2 Ab (<4 U)FPIR (mU/L)
11 3,4;2,8 Sibling 63 82 75 15 93 
48 3,4;2,7 Mother 160 92 89 −14 117 
44 3,12;2,7 Mother 160 −15 −1 260 
4,8;4,8 Sibling 20 760 49 100 62 
15 4,4;8,8 Sibling 75 120 42 88 234 
16 4,7;2,8 Sibling 25 3800 40 23 152 
15 4,4;8,8 Sibling 750 400 29 20 121 
16 3,3;2,2 Sibling 160 30 88 41 
46 4,4;8,8 Mother 129 90 49 89 44 
10 50 3,4;2,7 Mother 43 100 14 11 28 
11 14 2,4;1,8 Sibling 16 200 83 −5 151 
12 18 4,4;7,8 Sibling 50 44 86 −7 36 
a

ICA, islet cell Abs; JDF, Juvenile Diabetes Foundation; IAA, insulin autoantibodies; GAD Ab, Abs to glutamic acid decarboxylase; IA-2 Ab, Abs to tyrosine phosphatase IA-2; FPIR, first-phase insulin release, the sum of 1- and 3-min serum insulin concentrations after i.v. glucose injection; the threshold for the first percentile in a young adult population is 50 mU/L.

Table II.

Properties of DC generated from patients with recently diagnosed IDDMa

SubjectAgeSexHLADC Yield/107 PBMC% PositiveMFIMean ± SD × 10−3 cpm
DRDQ(×10−5)B7-1B7-2CD1aDRB7-1B7-2CD1aDRAllo 10,000Allo 2,000Auto 10,000Auto 2,000
IDDM 1 40 3,4 2,8 5.0 22 36 92 80 460 569 243 320 56.9 ± 2.55 35.6 ± 8.11 9.29 ± 0.29 8.38 ± 1.97 
Control 1 52 3,4 2,8 7.0 78 72 99 88 625 636 799 641 68.6 ± 5.01 53.9 ± 12.0 62.9 ± 5.44 39.9 ± 3.27 
IDDM 2 33 3,4 2,8 5.5 59 74 98 91 515 200 708 483 80.2 ± 8.11 75.2 ± 4.97 1.91 ± 2.74 4.32 ± 2.1 
Control 2 35 3,4 2,8 8.0 83 58 93 99 557 646 739 660 51.4 ± 12.3 61.2 ± 8.31 54.8 ± 1.32 57.8 ± 10.8 
IDDM 3 47 3,4 2,8 3.6 34 30 74 96 524 633 630 693 27.6 ± 4.08 55.3 ± 6.03 64.2 ± 3.90 38.4 ± 3.09 
Control 3 47 3,4 2,8 12.6 69 84 97 96 581 700 758 714 87.4 ± 5.06 79.5 ± 14.9 101 ± 9.66 79.5 ± 2.83 
SubjectAgeSexHLADC Yield/107 PBMC% PositiveMFIMean ± SD × 10−3 cpm
DRDQ(×10−5)B7-1B7-2CD1aDRB7-1B7-2CD1aDRAllo 10,000Allo 2,000Auto 10,000Auto 2,000
IDDM 1 40 3,4 2,8 5.0 22 36 92 80 460 569 243 320 56.9 ± 2.55 35.6 ± 8.11 9.29 ± 0.29 8.38 ± 1.97 
Control 1 52 3,4 2,8 7.0 78 72 99 88 625 636 799 641 68.6 ± 5.01 53.9 ± 12.0 62.9 ± 5.44 39.9 ± 3.27 
IDDM 2 33 3,4 2,8 5.5 59 74 98 91 515 200 708 483 80.2 ± 8.11 75.2 ± 4.97 1.91 ± 2.74 4.32 ± 2.1 
Control 2 35 3,4 2,8 8.0 83 58 93 99 557 646 739 660 51.4 ± 12.3 61.2 ± 8.31 54.8 ± 1.32 57.8 ± 10.8 
IDDM 3 47 3,4 2,8 3.6 34 30 74 96 524 633 630 693 27.6 ± 4.08 55.3 ± 6.03 64.2 ± 3.90 38.4 ± 3.09 
Control 3 47 3,4 2,8 12.6 69 84 97 96 581 700 758 714 87.4 ± 5.06 79.5 ± 14.9 101 ± 9.66 79.5 ± 2.83 
a

Allo, allogeneic MLR; Auto, autologous MLR; 10,000 or 2,000 is the number of DC present in the allogeneic or autologous MLR.

Islet cell Abs were measured by indirect immunofluorescence with doubling dilutions of serum on frozen sections of pancreas from a blood group O donor, and results were calibrated to Juvenile Diabetes Foundation (JDF) units against a standard serum. Insulin autoantibodies were measured by a fluid phase 125I-labeled insulin binding-precipitation assay. Glutamic acid decarboxylase Abs and tyrosine phosphatase IA-2 Abs were measured by precipitation of [35S]methionine-labeled recombinant proteins synthesized after transcription-translation in the TnT-coupled reticulocyte lysate system (Promega, Madison, WI). The four Ab assays have been included in all Immunology of Diabetes Workshops and Proficiency Programs (sponsored by Juvenile Diabetes Foundation International).

PBMC were cultured in human tonicity RPMI 1640 containing 20 mM HEPES and 10% FCS. Recombinant human GM-CSF (rhGM-CSF) was purchased from PeproTech (Rocky Hill, NJ) or kindly provided by Dr. Nicos Nicola (The Walter and Eliza Hall Institute of Medical Research, Victoria, Australia). Recombinant human IL-4 (rhIL-4) was kindly provided by Schering Plough (Kenilworth, NJ).

PBMC were isolated from heparinized venous blood on Ficoll-Paque, resuspended in medium, and allowed to adhere to six-well plates (Costar, Cambridge, MA) coated with rat tail collagen. After 2 h at 37°C in 5% CO2, the nonadherent cells were removed by a very gentle rinse, and the adherent cells were cultured for 7 days in medium containing 600 U/ml rhGM-CSF and 400 U/ml rhIL-4.

Cells from all subjects were surface phenotyped by labeling with a panel of mouse mAbs (Table III) and analyzed on a FACScan (Becton Dickinson, Mountain View, CA). Cells were gated on forward and side scatter and by propidium iodide staining, and up to 10,000 cells with high forward and side scatter were counted. Data acquisition was performed with the FACScan Research Software, which automatically calculates mean fluorescence intensity (MFI) after setting markers on positive and negative peaks.

Table III.

Mouse mAbs used for phenotypic analysis of DC

CD Classification SpecificitymAb (Isotype)Sourcea
CD1a OKT6 (IgG1) ATCC 
CD14 MφP9 (IgG2b) BD 
CD54 84H10 (IgG1) Dr. Andrew Boyd 
CD58 TS2/9.1.4.3 (IgG1) ATCC 
CD80 MAB104 (IgG1) Immunotech 
CD86 33401A (IgG1) PharMingen 
HLA DR L243 (IgG2a) ATCC or BD 
CD Classification SpecificitymAb (Isotype)Sourcea
CD1a OKT6 (IgG1) ATCC 
CD14 MφP9 (IgG2b) BD 
CD54 84H10 (IgG1) Dr. Andrew Boyd 
CD58 TS2/9.1.4.3 (IgG1) ATCC 
CD80 MAB104 (IgG1) Immunotech 
CD86 33401A (IgG1) PharMingen 
HLA DR L243 (IgG2a) ATCC or BD 
a

Source: BD, Becton Dickinson; Dr. Andrew Boyd (Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia); Immunotech (Marseilles, France); PharMingen (San Diego, CA); ATCC, American Type Culture Collection (Manassas, VA).

Responder CD4 cells (1 × 105), positively selected with magnetic beads (MACS; Miltenyi Biotech, Bergisch Gladbach, Germany), were cultured in 96-well round-bottom microplates with different numbers of irradiated (3000 rad, 137Cs source) stimulator cells (DC or PBMC). Proliferation was measured on day 5 by a 6-h pulse with [3H]thymidine (0.5 μCi/well, Amersham Life Science, Buckingham, U.K.)

Responder PBMC (2 × 105) were cultured in 96-well round-bottom microplates with different numbers of irradiated (3000 rad, 137Cs source) DC. Proliferation was measured on day 5 by a 6-h pulse with [3H]thymidine.

Differences between groups were analyzed by the Mann-Whitney U test.

When adherent cells from PBMC were cultured with GM-CSF + IL-4, veiled cell aggregates appeared and increased rapidly in size over 2 to 4 days. Over the next 3 to 5 days, these aggregates became gradually less adherent and released large cells with multiple fine dendrites. The phenotypic and functional properties of these DC derived from monocytes in the presence of GM-CSF and either IL-4 or IL-7 were recently described (17). The proportion of monocytes in PBMC (12.1–17.7) was similar in IDDM relatives, IDDM patients, and controls, and the adherent cells and DC derived from them did not differ morphologically between the groups. However, the yield of DC (mean ± SD) from IDDM relatives was markedly reduced (7.8 ± 3.2 × 105/107 PBMC) compared with paired controls (17 ± 5.1 × 105, p = 0.0002; Fig. 1). Similarly, the yield of DC from each IDDM patient was reduced compared with control (Table II).

FIGURE 1.

Impaired yield of DC in IDDM relatives. PBMC were isolated on Ficoll-Paque, resuspended in RPMI 1640 medium, and allowed to adhere to six-well plates coated with rat tail collagen. After 2 h at 37°C in 5% CO2, the nonadherent cells were removed by a very gentle rinse, and the adherent cells were cultured for up to 7 days in medium containing 600 U/ml rhGM-CSF and 400 U/ml rhIL-4. DC were counted microscopically, with trypan blue exclusion of dead cells. Dotted lines join at-risk IDDM relatives and age-, sex-, and HLA-matched control subjects. Short horizontal lines indicate mean values.

FIGURE 1.

Impaired yield of DC in IDDM relatives. PBMC were isolated on Ficoll-Paque, resuspended in RPMI 1640 medium, and allowed to adhere to six-well plates coated with rat tail collagen. After 2 h at 37°C in 5% CO2, the nonadherent cells were removed by a very gentle rinse, and the adherent cells were cultured for up to 7 days in medium containing 600 U/ml rhGM-CSF and 400 U/ml rhIL-4. DC were counted microscopically, with trypan blue exclusion of dead cells. Dotted lines join at-risk IDDM relatives and age-, sex-, and HLA-matched control subjects. Short horizontal lines indicate mean values.

Close modal

Up-regulation of the MHC class II molecule, HLA-DR, and the MHC class I-like molecule, CD1a, and down-regulation of the monocyte marker, CD14, characterizes the adherent monocyte-derived DC (14, 15, 16, 17). The DRhighCD14−/low phenotype was displayed by 90 ± 8.8% (mean ± SD) of DC generated from IDDM relatives and 95 ± 3.6% from controls (Fig. 2,A). However, compared with controls, a significantly lower proportion of cells from IDDM relatives expressed CD1a (72 ± 20 vs 90 ± 11%, p = 0.01; Fig. 2,B) and the costimulator ligands CD80 or B7-1 (36 ± 25 vs 71 ± 11%, p = 0.001: Fig. 2,C) and CD86 or B7-2 (34 ± 24 vs 62 ± 22%, p = 0.02; Fig. 2 D). The proportions of cells that expressed CD54 or ICAM-1 (81 ± 23 vs 92 ± 10%) and CD58 or LFA-3 (76 ± 28 vs 89 ± 13%) were not significantly different. The only IDDM relatives in whom the proportion of B7-1-positive cells was not significantly decreased compared with HLA class II-matched controls were the four who were DR4,4;DQ7 or 8 homozygous. Otherwise, there were no apparent HLA associations.

FIGURE 2.

Altered phenotype of DC in IDDM relatives. DC were phenotyped by surface labeling with a panel of mouse mAbs (Table III) and analyzed by flow cytometry. A live gate was set by forward and side scatter and propidium iodide staining. Five to ten thousand cells with high forward scatter and side scatter were counted. Dotted lines join at-risk IDDM relatives and age-, sex-, and HLA-matched control subjects. Short horizontal lines indicate mean values.

FIGURE 2.

Altered phenotype of DC in IDDM relatives. DC were phenotyped by surface labeling with a panel of mouse mAbs (Table III) and analyzed by flow cytometry. A live gate was set by forward and side scatter and propidium iodide staining. Five to ten thousand cells with high forward scatter and side scatter were counted. Dotted lines join at-risk IDDM relatives and age-, sex-, and HLA-matched control subjects. Short horizontal lines indicate mean values.

Close modal

The expression levels of both B7-1 and B7-2 per cell, measured as MFI units (mean ± SD), were significantly lower in IDDM relatives than in controls (B7-1, 390 ± 195 vs 537 ± 103, p = 0.02; B7-2, 482 ± 148 vs 586 ± 157, p = 0.01). However, the levels of HLA-DR (89.9 ± 8.8 vs 95.5 ± 3.6) and CD1a (654 ± 119 vs 689 ± 74) were no different. The differences in B7-1 and B7-2 expression can be clearly seen in representative examples (Fig. 3, A and B) of histograms overlaid from IDDM relatives and paired control subjects.

FIGURE 3.

Impaired expression of B7-1 and B7-2 on DC from IDDM relatives. B7-1 (A) and B7-2 (B) expression was analyzed by flow cytometry as described in the Figure 2 legend. Overlapping histograms of MFI for representative IDDM relatives (black) numbered as in Table I and paired control subjects (gray) are shown, together with the negative (IgG1 isotype) control.

FIGURE 3.

Impaired expression of B7-1 and B7-2 on DC from IDDM relatives. B7-1 (A) and B7-2 (B) expression was analyzed by flow cytometry as described in the Figure 2 legend. Overlapping histograms of MFI for representative IDDM relatives (black) numbered as in Table I and paired control subjects (gray) are shown, together with the negative (IgG1 isotype) control.

Close modal

Autologous CD4 T cells displayed a significantly lower proliferative response to DC from IDDM relatives than from controls at stimulator:responder ratios of 1:10 to 1:50. For example, at 1:10 (104 DC, 105 CD4 T cells), [3H]thymidine uptake (cpm) for IDDM relatives vs controls was 27,596 ± 14,691 vs 42,308 ± 10,893 (p = 0.04) (Fig. 4,A). The response of allogeneic PBMC (from an HLA-DR 15,8;DQ2,7 individual) to DC from IDDM relatives compared with controls was decreased, but not significantly (Fig. 4 B).

FIGURE 4.

Impaired Ag-presenting capacity of DC to autologous CD4 T cells in IDDM relatives. Autologous CD4 cells (1 × 105) positively selected with magnetic beads (A) or allogeneic PBMC (2 × 105) from one individual (B) were cultured in 96-well round-bottom microplates with 1 × 104 irradiated (3000 rad, 137Cs source) stimulator DC. Proliferation was measured on day 5 by a 6-h pulse with [3H]thymidine. Dotted lines join at-risk IDDM relatives and age-, sex-, and HLA-matched control subjects. Short horizontal lines indicate mean values.

FIGURE 4.

Impaired Ag-presenting capacity of DC to autologous CD4 T cells in IDDM relatives. Autologous CD4 cells (1 × 105) positively selected with magnetic beads (A) or allogeneic PBMC (2 × 105) from one individual (B) were cultured in 96-well round-bottom microplates with 1 × 104 irradiated (3000 rad, 137Cs source) stimulator DC. Proliferation was measured on day 5 by a 6-h pulse with [3H]thymidine. Dotted lines join at-risk IDDM relatives and age-, sex-, and HLA-matched control subjects. Short horizontal lines indicate mean values.

Close modal

Monocyte-derived DC from three patients with recently diagnosed IDDM (Table II) displayed similar properties to DC from at-risk IDDM relatives. A lower proportion expressed B7-1 (38 ± 19 vs 76 ± 7%) and B7-2 (47 ± 24 vs 71 ± 13%), but not CD1a (88 ± 13 vs 97 ± 3%). In particular, they displayed decreased expression (MFI) of B7-1 (500 ± 35 vs 588 ± 35, p < 0.05) and B7-2 (477 ± 245 vs 661 ± 34, p < 0.05) and a decrease in the autologous MLR (e.g., 17,026 ± 18,598 vs 59,076 ± 19,831 cpm, p < 0.05 at 2 × 103 DC stimulators:105 CD4 T cells).

This is the first demonstration of DC abnormalities in humans with preclinical autoimmune disease. At-risk IDDM relatives with underlying islet autoimmunity but with no metabolic dysfunction of diabetes had impaired yield, phenotype, and function of DC generated from peripheral blood adherent cells in the presence of GM-CSF and IL-4. Expression of B7-1 and B7-2 costimulator molecules was decreased on DC from IDDM relatives, together with the autologous MLR in response to these DC. Although impairment of CD4 T cell function in IDDM relatives was not directly excluded in this study, we think this is an unlikely explanation for the impaired autologous MLR. First, DC are unique in their ability to stimulate T cells in the autologous MLR (18), and their decrease in B7 expression would provide an explanation for the impaired autologous MLR. Second, a significant defect in T cell function was not revealed in the allogeneic MLR. The allogeneic stimulus is strong, and the response might be less affected by changes in DC phenotype such as a decrease in the expression of B7. Third, we (19) and others (20, 21) have found that T cell responses to both islet and nonislet Ags in preclinical at-risk relatives and patients with recently diagnosed IDDM are increased, not decreased.

Because MHC molecules have a direct role in Ag presentation, as well as other possible indirect effects on DC function, we carefully matched relatives with MHC class II identical controls. We found no difference in the expression of HLA-DR between relatives and controls. Therefore, MHC class II molecules can probably be eliminated as a variable that accounts for the depressed autologous MLR in relatives. DC phenotype and function varied considerably between individuals, but heterogeneity of phenotype and function within groups did not appear to be related to HLA class II phenotype. However, all subjects shared at least two HLA risk alleles for IDDM, and the number of alleles or haplotypes was insufficient for statistical analysis.

DC generated from patients with recently diagnosed IDDM displayed similar changes to those from at-risk relatives, consistent with the view that these changes are associated with the underlying autoimmune disorder that leads to β cell destruction. The question arises whether the changes in DC are the cause or effect of islet autoimmunity. HLA DR and DQ matching at least suggests that impaired DC function is not due to genetic susceptibility at MHC class II loci, but whether impaired DC phenotype and function is a genetic or acquired trait could be answered only by studying identical twins or HLA identical sibling pairs that are discordant for islet autoimmunity. Nevertheless, impairment of DC function to present self-peptides on MHC class II molecules, reflected in the autologous MLR, may contribute to the development of autoimmune disease. In the first place, DC present self-peptides to nascent T cells in the thymus and mediate the process of negative selection that deletes high affinity, self-reactive T cells (6). If adult peripheral blood adherent cell-derived DC were to functionally mirror thymic DC, then negative selection of high affinity autoreactive T cells might be impaired and predispose to autoimmune disease. The Ag specificity of this process would be dictated by the MHC repertoire and not necessarily be restricted to IDDM autoantigens. Controlled studies should be undertaken to investigate whether DC phenotype and function are also impaired in humans at risk for other autoimmune diseases, although apart from IDDM, identification of individuals with preclinical disease remains problematic.

In addition to mediating deletion of autoreactive T cells, DC may also elicit tolerance in the periphery by activating autoregulatory T cells. In the autologous MLR, CD4 T cells that proliferate in response to self-MHC class II-peptide complexes on DC (22, 23) have been shown to have several effector functions, including immunosuppression in vitro (23, 24, 25). Furthermore, it has been known for many years that the autologous MLR is depressed in autoimmune disease-susceptible mice (26), including nonobese diabetic (NOD) mice susceptible to IDDM (27, 28), and in humans (29, 30) with IDDM. These observations are consistent with the idea that impairment of DC function in autoimmune disease leads to deficient generation of regulatory T cells. Because autoantigen-specific T cells can be detected in healthy individuals, autoimmune disease can be viewed ultimately as a failure of peripheral regulatory mechanisms. The NOD mouse is characterized not only by a low autologous MLR (27, 28) but by decreased sensitivity of myeloid progenitor cells to GM-CSF (31) and by a defect in bone marrow-derived APC function (32). Clare-Salzler et al. (7) found that when young NOD mice were given a single s.c. injection of DC purified from pancreatic but not other lymph nodes of adult NOD mice, their incidence of diabetes was significantly reduced. Pooled lymph node cells from the recipient mice were then shown to suppress the adoptive transfer of diabetes by splenic T cells from diabetic mice. This implies that the transfer of pancreatic lymph node DC, presumably loaded with islet Ag(s), induced regulatory cells. Recent studies have identified both CD4 (33, 34) and CD8 (35) regulatory T cells in NOD mice, generated by the mucosal administration of islet Ags (insulin, glutamic acid decarboxylase) that are associated with Th2 cytokine profiles and inhibit adoptive transfer of diabetes. CD1, an MHC class I-like molecule, has been implicated as a ligand for two different types of regulatory T cells, mucosal intraepithelial CD8 T cells in oral tolerance (36) and NK1+ T cells that secrete the Th2 cytokine IL-4 (37). CD8 γδ T cells that suppress diabetes in the NOD mouse are induced by administration of aerosol insulin to the mucosa (35), and NK1+ T cells are deficient in NOD mice (38). In IDDM relatives, the expression of CD1a per cell, in contrast to B7-1 and B7-2, was normal, but the yield of CD1a+ cells generated was decreased. Whether regulatory T cells are induced by Ag presentation on CD1 or other molecules that may be altered on DC, including mucosal DC, is a question that must be addressed.

At a molecular level, how could DC fail to efficiently generate regulatory T cells and thereby promote autoimmune disease? T cell responses through the T cell receptor are regulated by B7 molecules in a manner that is complex and still controversial. Impaired ligation of CD28 on T cells to B7-1 and B7-2 on DC may lead to suboptimal expression of T cell molecules required for T cell clonal expansion (IL-2R, IL-2, and CD40 ligand) and viability (Bcl-xL) (39). A variety of experimental models have shown that costimulation through CD28 is necessary for priming of Th2 cells, without which T cells default to the Th1 subset (40, 41, 42, 43, 44). Decreased expression of B7 molecules may therefore impair the generation of Th2 cells and shift the immune balance toward Th1, thereby promoting cell-mediated disease such as IDDM. In fact, progression of diabetes in the NOD mouse was accelerated in the presence of a CD28 deficiency (43). The situation is unlikely to be this simple, however. For instance, the differential interaction of B7-1 vs B7-2 is reported to influence the nature of the T cell response, with B7-2 promoting Th2 responses (8, 45). Furthermore, T cell activation is followed by up-regulation of CTLA-4, which also ligates to B7-1 and B7-2, but with significantly higher affinity than CD28 (46). Therefore, decreased expression of B7 molecules could favor interaction with CTLA-4 over CD28 on activated T cells. How signaling through CD28 and CTLA-4 is normally integrated and the effect of its perturbation on T cell function are not fully understood. In purified mouse T cells, CD28 and CTLA-4 deliver opposing positive and negative signals, respectively (47), although the claim that CTLA-4 is solely a negative regulator has been challenged (48). In any event, as well as decreased B7 signaling through CD28, the differential interaction of B7 with CTLA-4 might also impair generation of Th2 cells with regulatory properties.

In concluding, our findings in humans with islet autoimmunity would support the hypothesis that a defect in DC function, in the thymus to reduce editing of self-reactive T cells or in the periphery to impair induction of regulatory T cells, predisposes to IDDM.

We thank Dr. Peter Colman for islet Ab assays, Dr. Brian Tait for HLA typing, and Mrs. Margaret Thompson for secretarial assistance. Drs. Bill Heath and Andrew Lew made constructive comments on the manuscript.

1

K.T. was supported by Manpei Suzuki Diabetes Foundation International, and M.C.H. and L.C.H. by the National Health and Medical Research Council of Australia and the Angelo and Susan Alberti Program Grant from Juvenile Diabetes Foundation International. VicHealth has generously supported the Melbourne Pre-Diabetes Family Study from which at-risk IDDM relatives were recruited.

3

Abbreviations used in this paper: IDDM, insulin-dependent diabetes mellitus; DC, dendritic cells; GM-CSF, granulocyte/macrophage-CSF; rh, recombinant human; MFI, mean fluorescence intensity; NOD, nonobese diabetic.

1
Harrison, L. C., I. L. Campbell, P. G. Colman, N. Chosich, T. W. H. Kay, B. Tait, R. K. Bartholomeusz, H. DeAizpurua, J. L. Joseph, S. Chu, W. E. Kielczynski.
1990
. Type 1 diabetes: immunopathology and immunotherapy.
Adv. Endocrinol. Metab.
1
:
35
2
Honeyman, M. C., D. S. Cram, L. C. Harrison.
1993
. Glutamic acid decarboxylase 67-reactive T cells: a marker of insulin-dependent diabetes.
J. Exp. Med.
177
:
535
3
Rudy, G., N. Stone, L. C. Harrison, P. G. Colman, P. McNair, V. Brusic, M. B. French, M. C. Honeyman, B. Tait, A. M. Lew.
1995
. Similar peptides from two β cell autoantigens, proinsulin and glutamic acid decarboxylase, stimulate T cells of individuals at risk for insulin-dependent diabetes.
Mol. Med.
1
:
625
4
Durinovic-Bello, I., M. Hummel, A. G. Ziegler.
1996
. Cellular immune responses to diverse islet cell antigens in IDDM.
Diabetes
45
:
795
5
Roep, B. O..
1996
. T-cell responses to autoantigens in IDDM: the search for the holy grail.
Diabetes
45
:
1147
6
Sprent, J., S. R. Webb.
1995
. Intrathymic and extrathymic clonal deletion of T cells.
Curr. Opin. Immunol.
7
:
196
7
Clare-Salzler, M. J., J. Brooks, A. Chai, K. Van Herle, C. Anderson.
1992
. Prevention of diabetes in nonobese diabetic mice by dendritic cell transfer.
J. Clin. Invest.
90
:
741
8
Kuchroo, V. K., M. P. Das, J. A. Brown, A. M. Ranger, S. S. Zamvil, R. A. Sobel, H. L. Weiner, N. Nabavi, L. H. Glimcher..
1995
. B7-1 and B7-2 costimulatory molecules activate differentially the Th1/Th2 developmental pathways: application to autoimmune disease therapy.
Cell
80
:
707
9
Steptoe, R. J., A. W. Thomson.
1996
. Dendritic cells and tolerance induction.
Clin. Exp. Immunol.
105
:
397
10
Finkelman, F. D., A. Lees, R. Birnbaum, W. C. Gause, S. C. Morris.
1996
. Dendritic cells can present antigen in vivo in a tolerogenic or immunogenic fashion.
J. Immunol.
157
:
1406
11
Marth, T., W. Strober, R. A. Seder, B. L. Kelsall.
1997
. Regulation of transforming growth factor-β production by interleukin-12.
Eur. J. Immunol.
27
:
1213
12
Kalinski, P., C. M. Hilkens, A. Snijders, F. G. Snijewint, M. L. Kapsenberg.
1997
. IL-12-deficient dendritic cells, generated in the presence of prostaglandin E2, promote type 2 cytokine production in maturing human naive T helper cells.
J. Immunol.
159
:
28
13
Jansen, A., M. van Hagen, H. A. Drexhage.
1995
. Defective maturation and function of antigen-presenting cells in type 1 diabetes.
Lancet
345
:
491
14
Romani, N., S. Gruner, D. Brang, E. Kampgen, A. Lenz, B. Trockenbacher, G. Konwalinka, P. O. Fritsch, R. M. Steinman, G. Schuler.
1994
. Proliferating dendritic cell progenitors in human blood.
J. Exp. Med.
180
:
83
15
Sallusto, F., A. Lanzavecchia.
1994
. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor α.
J. Exp. Med.
179
:
1109
16
Zhou, L. J., T. F. Tedder.
1996
. CD14+ blood monocytes can differentiate into functionally mature CD83+ dendritic cells.
Proc. Natl. Acad. Sci USA
93
:
2588
17
Takahashi, K., M. C. Honeyman, L. C. Harrison.
1997
. Dendritic cells generated from human blood in granulocyte macrophage-colony stimulating factor and interleukin-7.
Hum. Immunol.
55
:
103
18
Nussenzweig, M. C., R. M. Steinman.
1980
. Contribution of dendritic cells to stimulation of the murine syngeneic mixed leukocyte reaction.
J. Exp. Med.
151
:
1196
19
Honeyman, M. C., N. Stone, H. DeAizpurua, M. J. Rowley, L. C. Harrison.
1997
. High T-cell responses to the glutamic acid decarboxylase (GAD) isoform 67 reflect a hyperimmune state that precedes the onset of insulin-dependent diabetes.
J. Autoimmun.
10
:
165
20
Petersen, L. D., G. Duinkerken, G. J. Bruining, R. A. W. van Lier, R. R. P. de Vries, B. O. Roep.
1996
. Increased numbers of in vivo activated T cells in patients with recent onset insulin-dependent diabetes mellitus.
J. Autoimmun.
9
:
731
21
Kallmann, B. A., M. Hüther, M. Tubes, J. Feldkamp, J. Bertrams, F. A. Gries, E. F. Lampeter, H. Kolb.
1997
. Systemic bias of cytokine production toward cell-mediated immune regulation in IDDM and toward humoral immunity in Graves’ disease.
Diabetes
46
:
237
22
Glimcher, L. H., D. L. Longo, I. Green, R. H. Schwartz.
1981
. Murine syngeneic mixed lymphocyte response. I. Target antigens are self Ia molecules.
J. Exp. Med.
154
:
1652
23
Smith, J. B., R. P. Knowlton.
1979
. Activation of suppressor T cells in human autologous mixed lymphocyte culture.
J. Immunol.
123
:
419
24
James, S. P., G. G. Yenokida, A. S. Graeff, C. O. Elson, W. Strober.
1981
. Immunoregulatory function of T cells activated in the autologous mixed lymphocyte reaction.
J. Immunol.
127
:
2605
25
Yamashita, U., S. Ono, H. Nakamura.
1982
. The syngeneic mixed leukocyte reaction in mice. II. The I region control of suppressor T cell activity induced in the syngeneic mixed leukocyte reaction.
J. Immunol.
128
:
1010
26
Hom, J. T., N. Talal.
1982
. Decreased syngeneic mixed lymphocyte reaction in autoimmune susceptible mice.
Scand. J. Immunol.
15
:
195
27
Serreze, D. V., E. H. Leiter.
1988
. Defective activation of T suppressor cell function in nonobese diabetic mice.
J. Immunol.
140
:
3801
28
Baxter, A., M. A. Adams, T. M. Mandel.
1989
. Comparison of high- and low-diabetes-incidence NOD mouse strains.
Diabetes
38
:
1296
29
Chandy, K. G., A. M. Charles, A. Kershnar, B. Buckingham, N. Waldeck, S. Gupta.
1984
. Autologous mixed lymphocyte reaction in man: XV. Cellular and molecular basis of deficient autologous mixed lymphocyte response in insulin-dependent diabetes mellitus.
J. Clin. Immunol.
4
:
424
30
Räsänen, L., H. Hyöty, M. Lehto, O.-P. Kallioniemi, J. Antonen, T. Huupponen, J. Karjalainen, P. Leiinikki.
1988
. Defective autologous mixed leucocyte reaction in newly diagnosed type 1 diabetes mellitus.
Clin. Exp. Immunol.
71
:
470
31
Langmuir, P. B., M. M. Bridgett, A. L. M. Bothwell, I. N. Crispe.
1993
. Bone marrow abnormalities in the non-obese diabetic mouse.
Int. Immunol.
5
:
169
32
Serreze, D. V., H. R. Gaskins, E. H. Leiter.
1993
. Defects in the differentiation and function of antigen-presenting cells in NOD/Lt mice.
J. Immunol.
150
:
2534
33
Bergerot, I., N. Fabien, V. Maguer, C. Thivolet.
1994
. Oral administration of human insulin to NOD mice generates CD4+ T cells that suppress adoptive transfer of diabetes.
J. Autoimmun.
7
:
655
34
Tian, J., M. A. Atkinson, M. Clare-Salzler, A. Herschenfeld, T. Forsthuber, P. V. Lehman, D. L. Kaufman.
1996
. Nasal administration of glutamate decarboxylase (GAD65) peptide induces Th2 responses and prevents murine insulin-dependent diabetes.
J. Exp. Med.
183
:
1561
35
Harrison, L. C., M. Dempsey-Collier, D. Kramer, K. Takahashi.
1996
. Aerosol insulin induces CD8 γ δ T cells that suppress murine insulin-dependent diabetes.
J. Exp. Med.
272
:
1811
36
Panja, A., R. S. Blumberg, S. P. Balk, L. Mayer.
1993
. CD1d is involved in T cell-intestinal epithelial cell interactions.
J. Exp. Med.
178
:
1115
37
Bendelac, A., O. Lantz, M. E. Quimby, J. W. Yewdell, J. R. Bennink, R. R. Brutkiewicz.
1995
. CD1 recognition by mouse NK1+ T lymphocytes.
Science
268
:
863
38
Gombert, J. M., A. Herbelin, E. Tancrede-Bohin, M. Dye, C. Carnaud, J.-F. Bach.
1996
. Early quantitative and functional deficiency on NK1+-like thymocytes in the NOD mouse.
Eur. J. Immunol.
26
:
2989
39
Boussiotis, V. A., G. J. Freeman, J. G. Gribben, L. M. Nadler.
1996
. The role of B7-1/B7-2:CD28/CTLA-4 pathways in the prevention of anergy, induction of productive immunity and down-regulation of the immune response.
Immunol. Rev.
153
:
5
40
Corry, D. B., S. L. Reiner, P. S. Linsley, R. M. Locksley.
1994
. Differential effects of blockade of CD28–B7 on the development of Th1 or Th2 effector cells in experimental leishmaniasis.
J. Immunol.
153
:
4142
41
King, C.L., R. J. Stupi, N. Craighead, C. H. June, G. Thyphronitis.
1995
. CD28 activation promotes Th2 subset differentiation by human CD4+ cells.
Eur. J. Immunol.
25
:
587
42
Webb, L. M., M. Feldmann.
1995
. Critical role of CD28/B7 costimulation in the development of human Th2 cytokine-producing cells.
Blood
86
:
3479
43
Lenschow, D. J., K. C. Herold, L. Rhee, B. Patel, A. Koons, H. Y. Qin, R. Fuchs, B. Singh, C. B. Thompson, J.A. Bluestone.
1996
. CD28/B7 regulation of Th1 and Th2 subsets in the development of autoimmune diabetes.
Immunity
5
:
285
44
Ruflifson, I. C., A. I. Sperling, P. E. Fields, F. W. Fitch, J. A. Bluestone.
1997
. CD28 costimulation promotes the production of Th2 cytokines.
J. Immunol.
158
:
658
45
Freeman, G. J., V. A. Boussiotis, A. Anumanthan, G. M. Bernstein, X. Y. Ke, P. D. Rennert, G. S. Gray, J.G. Gribben, L. M. Nadler.
1995
. B7-1 and B7-2 do not deliver identical costimulatory signals, since B7-2 but not B7-1 preferentially costimulates the initial production of IL-4.
Immunity
2
:
523
46
van der Merwe, P. A., D. L. Bodian, S. Daenke, P. Linsley, S. J. Davis.
1997
. CD80 (B7-1) binds both CD28 and CTLA-4 with a low affinity and very fast kinetics.
J. Exp. Med.
185
:
393
47
Krummel, M. F., J. P. Allison.
1995
. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation.
J. Exp. Med.
182
:
459
48
Liu, Y..
1997
. Is CTLA-4 a negative regulator for T-cell activation?.
Immunol. Today
18
:
569