当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 糖尿病学杂志 > 2006年 > 第1期 > 正文
编号:11256904
The Diabetes Type 1 Locus Idd6 Modulates Activity of CD4+CD25+ Regulatory T-Cells
     1 Unitee de Geeneetique Moleeculaire Murine, Centre National de Recherche (CNRS) Unitee de Recherche Associeee (URA) 2578, Institut Pasteur, Paris, France

    2 Hpital Cochin St. Vincent de Paul, Paris, France

    FACS, fluorescence-activated cell sorter; IAA, insulin autoantibody; QTL, quantitative trait locus

    ABSTRACT

    The genetic locus Idd6 confers susceptibility to the spontaneous development of type 1 diabetes in the NOD mouse. Our studies on disease resistance of the congenic mouse strain NOD.C3H 6.VIII showed that Idd6 influences T-cell activities in the peripheral immune system and suggest that a major mechanism by which the Idd6 locus modifies diabetes development is via modulation of regulatory T-cell activities. Our transfer experiments using total splenocytes and purified T-cells demonstrated that the locus specifically controls the efficiency of disease protection mediated by the regulatory CD4+CD25+ T-cell subset. Our data also implicate the Idd6 locus in controlling the balance between infiltrating lymphocytes and antigen-presenting cells within the pancreatic islet.

    Over 20 type 1 diabetes susceptibility loci (Idd) have been genetically localized in the NOD mouse (1), but little information about the nature of these noneCmajor histocompatibility complex Idd genes has been obtained and few candidate genes have been proposed. The construction of congenic strains that differ from the NOD receiver strain by only a selected genetic region derived from a noneCdiabetes- prone parental donor strain (2,3) is a widely used approach to define disease-related candidate regions. All mice of a given congenic strain are genetically identical, which allows a wide variety of phenotypic studies to be applied to large groups of genetically uniform animals. A promising strategy for candidate gene identification is to combine phenotype analysis of congenic mice with expression profiling and haplotype and mutational analysis (4eC7).

    Recently, type 1 diabeteseCassociated regions on distal chromosome 6, the Idd6, Idd19, and Idd20 loci, have been further defined by the analysis of a series of congenic strains, carrying C3H/HeJ genomic material for distal chromosome 6 introgressed onto the NOD/Lt genetic background (8). NOD/Lt alleles at the Idd6 locus confer susceptibility to type 1 diabetes, whereas C57BL/6, C57BL/10, and C3H/HeJ alleles all confer resistance to diabetes (8eC10). The NOD.C3H congenic strain described in this study carries NOD alleles at both the Natural Killer gene complex (10) and the candidate region for the islet-specific BDC-6.9 autoantigen gene (11,12), which excludes both loci from being responsible for the disease resistance.

    Three observations have contributed to the idea that Idd6 might act via the control of T-cell activities: 1) that a quantitative trait locus (QTL) conferring resistance of NOD-derived immature CD4+/CD8+ thymocytes to dexamethasone-induced apoptosis maps to within the Idd6 region in a NOD x C57BL/6 cross (13eC15); 2) that Idd6 controls low rates of proliferation in immature NOD thymocytes (16); and 3) that the protective effects of C3H/HeJ alleles introgressed at the 4.5-cM Idd6 interval are completely abolished in NOD.C3H 6.VIII congenic mice after they are treated with cyclophosphamide, an alkylating agent that leads to the depletion of regulatory T-cells (17,18), leading us to speculate that the Idd6 locus controls the presence or activity of a population of suppressive T-cells in young mice (8).

    In the present study, we undertook a detailed phenotypic analysis of the congenic strain NOD.C3H 6.VIII (8), which shows resistance to the spontaneous development of diabetes. We have shown that this resistance is not attributable to either the intrinsic resistance of islet -cells to immune destruction or the defect in pathogenic T-cells. Protection of the congenic strain likely involves the control of the proportions of the different leukocyte subsets infiltrating the islet, and in particular that of CD4+ T-cells. Critical to the reduced diabetes susceptibility of the Idd6 congenic mice is our finding that their regulatory CD4+CD25+ T-cell subset confers enhanced disease protection.

    RESEARCH DESIGN AND METHODS

    The congenic strain NOD.C3H 6.VIII, homozygous for C3H alleles at the Idd6 locus; the control congenic strain, carrying NOD alleles at the Idd6 locus (8); and the NOD/Lt, NOD/scid, and C3H/HeJ mice were all maintained in our animal house by brother-sister mating. The NOD/scid.C3H 6.VIII strain (6.VIII/scid) was established from the congenic strain 6.VIII by crossing it to the NOD/scid strain. F1 generation mice were intercrossed, and mice homozygous for both the C3H-derived Idd6 interval (markers D6Mit14, 57, 15, 304) and the scid mutation were selected. The animal studies were approved by the institutional review boards.

    Histopathology of the pancreas.

    Pancreata were excised, fixed in Bouin’s solution, and processed for paraffin embedding. Then four 5-e蘭 sections taken at 100-e蘭 intervals were stained using hematein-eosin-safranin. At least 20 islets per specimen were analyzed.

    Immunofluorescence staining.

    Islet-infiltrating leukocytes were isolated as previously described (19). Cells were pelleted in 96-well plates and stained for 30 min on ice in 20 e蘬 of PBS supplemented with 2% FCS and 5 mmol/l sodium azide using reagents labeled for biotin, phycoerythrin, fluorescein isothiocyanate, peridinin chlorophyll, or allophyrocyanin (BD Bioscience, Le Pont de Claix, France) at optimal concentrations. Where appropriate, a secondary staining step using fluorochrome-conjugated streptavidin was performed. Cells were washed twice and resuspended in PBS containing 1% formaldehyde. Flow cytometry analysis was performed using a FACScalibur and CellQuest software (BD Biosciences, Grenoble, France). The sample size for data collection was 10,000 cells.

    Insulin autoantibody determination.

    The 96-well filtration plate microassay for insulin autoantibodies (IAAs) (20) was performed as previously described (21).

    Cell purification.

    To purify CD4+ cells, spleen cell suspensions were incubated on ice for 20 min with a mixture of biotin-conjugated anti-CD8, antieCMac-1, anti-Gr1, anti-B220, and TER-119 antibodies; washed; and then incubated with streptavidin microbeads (Milteny Biotec, Paris, France). Cells were separated using a VarioMACS device according to the manufacturer’s protocol. The nonretained cells (90eC95% CD4+) were further sorted using a biotinylated anti-CD25 (7D4) antibody and streptavidin microbeads. After cells were stained with fluorescein isothiocyanateeCanti-CD4 and phycoerythrineCanti-CD25 (PC61) antibodies, they were analyzed by fluorescence-activated cell sorter (FACS), which reproducibly showed that the retained cells were composed of 80% CD4+CD25+ cells. The negative fraction contained 90eC95% CD4+CD25eC cells. Magnetic cell sorting was used to enrich the latter cell population for CD4+CD25eCCD62L+ cells. In some experiments, CD4+CD62L+ cells were directly enriched from the CD4+ cell pool. Diabetogenic CD62LeCCD25eC T-cells (70eC80%) (22) were enriched by the depletion of B-cells, macrophages, erythroid cells, and CD62L+ and CD25+ cells by magnetic cell sorting using a pool of three to six spleens from diabetic mice or animals age 15 weeks.

    Adoptive transfer of diabetes.

    Cells were injected intravenously into immunodeficient recipients: NOD/scid mice, irradiated mice, or thymectomized irradiated mice. Thymic ablation was performed on 6-week-old mice under anesthesia (0.01 ml/g body wt of a 2.5% solution of Avertin [Aldrich]) by suction using a Pasteur pipette. To normalize diabetes transfers, the injected number of diabetogenic CD62LeCCD25eC T-cells was adjusted to 5 x 105 cells per recipient. In cotransfer experiments, NOD/scid recipients were injected with 5 x 105 CD62LeCCD25eC T-cells from diabetic mice together with adequate numbers of purified test population cells. Recipients were monitored for diabetes for 10eC12 weeks after cells were transferred by the assessment of glucosuria.

    Statistical analysis.

    Pooled data computed as means ± SE were compared using the Mann-Whitney test. Time-to-event distributions were calculated by Kaplan-Meier estimation and compared by log-rank tests over the period of observation.

    RESULTS

    Protection against diabetes but not against insulitis.

    The congenic strain NOD.C3H 6.VIII (6.VIII) was developed by backcrossing the C3H/HeJ-derived chromosome 6 interval distal to D6Mit57 onto the NOD/Lt genetic background. Over the 8-month test period, diabetes incidence was significantly reduced in congenic 6.VIII mice as compared with in control congenic mice in both female (P < 0.0001) and male (P = 0.0011) animals (Fig. 1A). To investigate if the decreased diabetes incidence correlated with changes in insulitis severity, we evaluated the extent of insulitis in pancreas sections obtained from pre-diabetic female strain 6.VIII and control mice (Fig. 1B). Neither 6.VIII nor control mice showed pancreatic islet abnormalities at age 4 weeks. The 8-week-old mice of both strains developed peri-islet and intraislet infiltration. Although a clear progression from peri-insulitis to invasive insulitis was observed with age, no difference in the extent of insulitis was observed between the two strains. Males of both groups developed less diabetes and generally milder insulitis than females. For example, in 16-week-old control male mice, the average percentage of peri-insulitis plus invasive insulitis was 48% versus 81% in female mice. The extent of sialitis development was also found to be similar in both strains (data not shown). These data indicate that C3H alleles at the Idd6 locus conferred protection against diabetes but not against pancreatic insulitis and sialitis.

    Presence of IAAs.

    Because the presence of IAAs has been correlated with the likelihood of diabetes development in the NOD mouse (20), we measured the IAA levels by radioimmunoassay in female mice of both the 6.VIII and the control strain at age 12 and 16 weeks. In 12-week-old mice, the percentage of animals positive for IAA was lower in the 6.VIII group than in the control group (P < 0.01); however, this difference was not observed at age 16 weeks (P > 0.05) (Fig. 2). These results suggest that the Idd6 locus does not control IAA levels, but that the lower IAA levels at age 12 weeks do correlate with the delay in diabetes development observed in 6.VIII mice.

    Reduced frequencies in islet-infiltrating lymphocytes.

    The absence of any difference in the extent of insulitis between the two strains prompted us to search for more subtle differences in the leukocyte subsets invading the islets. We analyzed the islet infiltrate of 12-week-old female mice by FACS. The frequency of infiltrating CD4+ and B-cells (P < 0.05) was significantly lower in 6.VIII islets than in control islets. The CD8+ cell subset, however, remained unchanged. The reduction in the number of lymphocytes infiltrating the 6.VIII islets was counterbalanced by an increase in the number of infiltrating nonlymphoid cells, including macrophages (CD11b+ and F4/80+; P < 0.05) and dendritic cells (CD11c+; P < 0.05) (Fig. 3). This change in the nature of islet-infiltrating cells might modulate the aggressiveness of the autoimmune response to -cells.

    To determine whether these changes were limited to cells in the pancreas, we performed FACS analysis of the splenic and thymic cell populations of control and 6.VIII mice using six 8-week-old females of each strain. We observed 43.8 ± 3.4 and 43 ± 4% B-cells, 10.7 ± 1.3 and 10.5 ± 1.8% CD8+ cells, and 29.5 ± 2.8 and 31.3 ± 3.9% CD4+ cells in the spleen of control and 6.VIII mice, respectively. The percentages of regulatory CD25+ T-cells within the CD4+ subset were also comparable: 10.6 ± 0.4 and 9.6 ± 0.4% in control and 6.VIII mice, respectively.

    In the thymus, the percentages of T-cell subpopulations in the control (CD4eCCD8eC, 3.4 ± 0.2; CD4+CD8+, 84 ± 1.7; CD4+, 9.3 ± 1; and CD8+, 3.5 ± 0.3%) and the 6.VIII (CD4eCCD8eC, 3.8 ± 0.3; CD4+CD8+, 83.3 ± 1.5; CD4+, 9.2 ± 0.5; and CD8+, 3.8 ± 0.4%;) strains were similar. The percentage of CD25+ cells within the CD4+ T-cell population was 3.9 ± 0.2 and 3.6 ± 0.4% in the control and 6.VIII strains, respectively. We concluded that the reduction in lymphocyte number in the pancreatic infiltrate of the diabetes-resistant strain 6.VIII compared with the control strain did not reflect more generalized changes in the cellular composition of the lymphoid system.

    Resistance of diabetes transfer requires the immune system.

    To investigate the hypothesis of a resistance of -cells to immune destruction, we tested the ability of diabetogenic NOD splenocytes to transfer diabetes to control and 6.VIII mice. The recipients were irradiated (750 rad) 1 day before 107 spleen cells were transferred from diabetic mice. As shown in Fig. 4A, female 6.VIII recipients were strongly protected against diabetes transfer, with only 33% becoming diabetic versus 80% of the control recipients 7 weeks after the transfer (P = 0.007). Males showed similar results, with a final diabetes incidence of 36% for 6.VIII (n = 11) and 79% for control (n = 14) animals.

    To determine if this protection was actually related to the susceptibility of -cells to destruction, we performed two additional experiments. First, mice were thymectomized at age 6 weeks, irradiated 1 week later, and injected with diabetogenic spleen cells the next day. As shown in Fig. 4B, thymectomized, irradiated 6.VIII recipients lost their protection against diabetes transfer. This suggests that the thymus is required to protect irradiated 6.VIII mice against diabetes transfer. In a second experiment, we constructed a novel congenic strain, NOD/scid.C3H 6.VIII (6.VIII/scid), that is homozygous for both the C3H/HeJ-derived Idd6 interval and the Prkdcscid mutation (23). Homozygous scid mice have been found to lack functional T- and B-cells. Splenocytes from diabetic NOD mice were transferred to 6-week-old male or female 6.VIII/scid and NOD/scid mice. All the scid recipients developed diabetes within 6eC7 weeks after the transfer, irrespective of whether they carried the C3H alleles at the Idd6 locus or not (Fig. 4C). At 4 weeks after the transfer, the extent of insulitis was comparable in each strain, with 35% of the islets showing peri-insulitis and 18% showing intra-insulitis (n = 4 recipients per strain). These results indicated that the genetic transfer of the C3H-derived Idd6 interval did not result the islets becoming resistant to autoimmune attack in the absence of the immune system.

    Presence and activity of diabetogenic T-cells.

    In light of the above experiments, the immune system, the thymus, and particularly T-cells appear to be crucial to the protection afforded by genes at the Idd6 locus. We then tested whether diabetogenic T-cells from 6.VIII mice have reduced activity as compared with those from control mice. In transfer experiments we used both total spleen cells and splenocytes enriched for CD25eCCD62LeC T-cells from aged mice. Both cell populations have been shown to concentrate diabetogenic activity (24,25), and CD25eCCD62LeC T-cells have been observed to be devoid of regulatory T-cells (22; F.L., M.-C.G., unpublished observations).

    Adoptive transfer of splenocytes to NOD/scid mice was performed using pre-diabetic 15-week-old male and female control and 6.VIII donors. Splenocytes from aged NOD mice are expected to induce diabetes rapidly in immunodeficient recipients due to the progressive decrease in the activity of regulatory T-cells and the enhanced pathogenicity of CD25eC T-cells with age (25,26). Aged pre-diabetic mice allow a better estimation of the diabetogenic T-cell pool size than diabetic mice because the latter most likely harbor equivalent numbers of effector T-cells independent of their genotype. Splenocytes from diabetic NOD mice were used as positive controls and allowed a rapid induction of diabetes within 3eC5 weeks after transfer. Diabetes developed with similar kinetics when donor cells of male and female pre-diabetic control and 6.VIII mice were used, with diabetes being observed from 6 weeks onwards (Fig. 5A). Our results suggest that splenocytes from aged 6.VIII mice induce diabetes as efficiently as cells from aged control mice, with both showing a delay of at least 3 weeks compared with splenocytes from diabetic NOD mice. This delay was likely associated with the use of nonpurified splenocytes in the transfer experiment, which contain both effector and persistent regulatory T-cells.

    Effector cells have been shown to express a memory/activated T-cell type phenotype. We therefore transferred 0.5 x 106 CD25eCCD62LeC purified T-cells (a number of diabetogenic cells equivalent to that contained in 107 total splenocytes) from 15-week-old control and 6.VIII male or female mice into 5 week-old male NOD/scid recipients (Fig. 5B). As in the previous experiment, diabetogenic cells from the four groups of 15-week-old mice induced diabetes with similar kinetics, although in this case, the progression was delayed for only 1eC2 weeks compared with in cells from diabetic mice (Fig. 5B). When the number of cells from pre-diabetic mice was increased from 0.5 x 106 cells to 106 cells, similar transfer kinetics were observed (Fig. 5C). We concluded that the protective effect of Idd6 on the onset of diabetes is unlikely to be based on differences in the presence or activity of pathogenic T-cells.

    Evidence for the control of regulatory T-cells by Idd6.

    Splenocytes from young, 8 week-old mice contain few diabetogenic cells and efficiently inhibit diabetes in cotransfer with diabetogenic cells (27,28). A total of 2 x 107 splenocytes isolated from mice of each strain were injected together with 107 splenocytes from diabetic NOD mice into recipient mice. Diabetes incidence was observed weekly for 10 weeks after the adoptive transfer. Compared with the transfer of diabetogenic cells alone, which induced diabetes in 90% of the recipients at 10 weeks, coinjection of splenocytes from 6.VIII mice conferred an almost complete protection against diabetes, with only 1 of 16 recipients becoming diabetic within 10 weeks (P < 0.0001). Co-injection of splenocytes from young control mice was partially protective (P = 0.005). The difference in the protection conferred by spleen cells from strain 6.VIII versus that from the control strain was statistically significant (P = 0.0014) (Fig. 6A). We concluded that both 6.VIII and control splenocytes contain suppressor cells that are able to control the development of diabetes, but that 6.VIII spleen cells exhibit significantly higher protective activity.

    Suppressor activity of strain 6.VIII regulatory T-cells.

    To better define the regulatory T-cells involved in the Idd6-dependent protective mechanism, we performed cotransfer experiments using purified T-cell subsets. In young pre-diabetic NOD mice, the CD4+ T-cell subset, originally reported as being able to control diabetes development (27), contains at least two populations of regulatory T-cells that express CD25+ and/or CD62L+ (28eC30). We first tested the regulatory potential of the CD4+CD62L+ T-cell population, which contains regulatory T-cells belonging to both the CD25+ and CD25eC subsets (F.L., M.-C.G. unpublished observations). Young NOD/scid mice were injected with 0.5 x 106 CD62LeCCD25eC diabetogenic cells together with 10 x 106 CD4+CD62L+ T-cells from 6- to 8-week-old mice. In full agreement with the results obtained when total splenocytes were coinjected (Fig. 6A), none of the five cotransfer recipients of 6.VIII CD4+CD62L+ splenocytes became diabetic. Less efficient protection was again observed when cells from control mice were used (P = 0.002 vs. the diabetogenic cell group) (Fig. 6B).

    In view of this result, the regulatory potential of CD4+CD25+ and CD4+CD25eCCD62L+ T-cell populations was tested independently. Young NOD/scid mice were injected with purified diabetogenic cells together with previously determined optimal cell numbers of either CD4+CD25+ or CD4+CD25eCCD62L+ T-cells from 6- to 8-week-old congenic mice (28,29). Our experiments showed that 2 x106 CD4+CD25+ cells from strain 6.VIII protected the recipients from diabetes transfer more efficiently than did the equivalent cells from the control strain (P < 0.05), whereas no differences were found comparing the two strains in cotransfer experiments using either 9 x106 (Fig. 6C) or 2 x106 CD4+CD25eCCD62L+ T-cells (data not shown).

    To determine whether this difference in inhibitory activity persists over time, we performed a cotransfer experiment using purified CD4+CD25+ and CD4+CD25eCCD62L+ regulatory T-cells from 15-week-old donors. Again, all four cell populations were found to be protective (P < 0.0005 for each cotransferred population vs. diabetogenic cells). CD4+CD25+ T-cells from both the 6.VIII and control strains provided efficient protection from diabetes transfer (P < 0.0001), with the greatest suppressive activity associated with 6.VIII cells (Fig. 6D). Again, no difference in protection was observed between 6.VIII CD4+CD25eCCD62L+ T-cells and control CD4+CD25eCCD62L+ T-cells (Fig. 6C and D). Although the suppressive activity of CD4+CD25+ cells of both strains persisted over time, only cells from 6.VIII mice were able to ensure complete protection over a 10-week period (P 0.05).

    DISCUSSION

    The mechanisms that trigger the activation of autoreactive lymphocytes directed against insulin-producing -cells are still largely unknown. Alteration of known diabetes-implicated genes, such as the MHC class II, the insulin, and the CTLA4 genes cannot, by itself, account for the development of type 1 diabetes. An association of subphenotypes, each controlled by a unique or several QTLs, that imprint a functional specificity on the key steps in the development of immune reactions or -cell functions likely underlies the development of diabetes.

    The present study focused on the analysis of the congenic NOD.C3H 6.VIII mouse strain. Both male and female congenic 6.VIII mice developed insulitis, although they were protected from diabetes. The 6.VIII strain exhibited reduced levels of anti-IAAs at age 12 weeks, which correlated with its degree of protection from spontaneous diabetes (20). The efficient transfer of diabetes and insulitis by splenocytes from diabetic NOD donors into 6.VIII/scid recipients, however, excludes the possibility that the diabetes resistance of this strain is due to intrinsic -cell modifications conferring major resistance to immune destruction. Moreover, the resistance of pre-irradiated 6.VIII recipients to the transfer suggests that the protection relies on a radio-resistant process that requires the host immune system.

    Our finding that the frequency of islet-infiltrating CD4+ T-cells and B-cells decreases in 6.VIII mice whereas the percentage of macrophages and dendritic cells increases underscores the possible role of the local infiltrate in disease protection. It can be hypothesized that a switch in the representation of lymphoid subsets within the islets is directly involved in diabetes protection and may be critical in determining whether the initial infiltrate proceeds to diabetes or whether steady-state insulitis can be maintained. In contrast to the infiltrate, no major change was found in the distribution of T-cell subsets in primary and secondary lymphoid organs in 6.VIII mice, suggesting that the QTL associated with the CD4-to-CD8 T-cell ratio between C57BL/6 and DBA/2 mice (Tmq1) (31) is unlikely to be controlled by NOD or C3H alleles at Idd6.

    The putative role of the immune system in the diabetes protection afforded by C3H alleles at Idd6 suggests two different hypotheses: decreased activity of effector cells or increased efficiency of immune regulatory mechanisms. The possibility of reduced activity of effector T-cells has been excluded, as diabetes transfer using diabetogenic cells from 6.VIII and control congenics gave comparable results. In contrast, we obtained clear evidence to support the hypothesis that regulatory T-cells are involved in the diabetes protection of 6.VIII mice. The first indication was provided by results showing that thymectomy abrogated the resistance of irradiated 6.VIII congenics to diabetes transfer. The thymus is known to be the major site for the generation of regulatory T-cells, and impairment of the regeneration of thymocytes has been shown to rapidly induce diabetes in the NOD model (27,32eC34). Co-transfer experiments using splenocytes from young 6.VIII mice have shown that protection from diabetes can be transferred by splenocytes from the disease-protected mice. Both purified CD4+CD25+ and purified CD4+CD62L+ T-cell subsets showed suppressive activity against the transfer of diabetes by effector T-cells from diabetic donors. In all the experiments that we have undertaken, the highest protective activity was associated with the CD4+CD25+ T-cell subset, with 6.VIII CD4+CD25+ T-cells showing significantly higher suppressive activity than control CD4+CD25+ T-cells. This phenotypic observation leads us to draw the qualitative conclusion that Idd6 alleles modulate the efficiency of CD4+CD25+ regulatory T-cells. The interactions of regulatory T-cells with other cell types likely play an important role in the protection conferred by C3H/HeJ alleles at the Idd6 locus. The local changes in the islet infiltrate suggest such interactions may take place in either the islet itself or the draining lymph nodes. Previous studies have shown that pancreatic lymph nodes are a major crossroads in which the immune response to -cells occurs in the NOD model (35,36). At present, it is not clear whether one or a combination of genes in the Idd6 interval control the different phenotypes that have been observed.

    The importance of regulatory T-cells in the control of autoimmunity, including the well-characterized CD4+ T-cells expressing the interleukin-2 receptor (CD25) (37,38) and L-selectin (CD62L) (28), has been known for a long time (27,39,40), and their inhibitory function on the activation and proliferation of pathogenic T-cells is well characterized (41). Recent work has clearly demonstrated the role of CD4+CD25+ T-cells in the development of type 1 diabetes (42). Defining a cell type whose activity is controlled by Idd6 in the NOD mouse has led us one step further toward identifying the molecular mechanisms underlying Idd6 action and the relevant disease genes lying within its candidate region. Phenotypes associated with regulatory T-cell activity may also contribute to the other genetic traits that localize to the Idd6 region, such as the susceptibility to systemic lupus erythematosus (Lbw4) (43) or to lung and skin cancer (44eC46). Because none of the genes known to be implicated in T-cell regulatory activity, such as the transcription factor forkhead box P3 (47eC49) or the tumor growth factor 1 (50), localize to the Idd6 interval, the definition of the Idd6 genes is likely to contribute in novel ways to our understanding of the genetic pathways that underpin regulatory T-cell action in controlling immune responses. The definition of a cellular phenotype correlating with Idd6 spontaneous resistance provides an important source of information for the transcriptional profiling approaches that, with sequence and polymorphism evaluation, underlie our current efforts to define and functionally test Idd6 candidate genes.

    ACKNOWLEDGMENTS

    This study was financed by research grants from the Juvenile Diabetes Research Foundation International (1-2000-600) and the Ministeere de la Recherche et Technologie (03246) and by funds from INSERM, the CNRS, and the Pasteur Institute.

    FOOTNOTES

    U.C.R. and F.L. contributed equally to this study.

    REFERENCES

    Deruytter N, Boulard O, Garchon HJ: Mapping non-class II H2-linked loci for type 1 diabetes in nonobese diabetic mice. Diabetes 53:3323eC3327, 2004

    Prochazka M, Serreze DV, Worthen SM, Leiter EH: Genetic control of diabetogenesis in NOD/Lt mice: development and analysis of congenic stocks. Diabetes 38:1446eC1455, 1989

    McAleer MA, Reifsnyder P, Palmer SM, Prochazka M, Love JM, Copeman JB, Powell EE, Rodrigues NR, Prins JB, Serreze DV, DeLarato NH, Wicker LS, Peterson LB, Schork NJ, Todd JA, Leiter EH: Crosses of NOD mice with the related NON strain: a polygenic model for IDDM. Diabetes 44:1186eC1195, 1995

    Wicker LS, Todd JA, Peterson LB: Genetic control of autoimmune diabetes in the NOD mouse. Annu Rev Immunol 13:179eC200, 1995

    Rogner UC, Avner P: Congenic mice: cutting tools for complex immune disorders. Nat Rev Immunol 3:243eC252, 2003

    Lyons PA: Gene-expression profiling and the genetic dissection of complex disease. Curr Opin Immunol 14: 627eC630, 2002

    Eckenrode SE, Ruan Q, Yang P, Zheng W, McIndoe RA, She JX: Gene expression profiles define a key checkpoint for type 1 diabetes in NOD mice. Diabetes 53:366eC375, 2004

    Rogner UC, Boitard C, Morin J, Melanitou E, Avner P: Three loci on mouse chromosome 6 influence onset and final incidence of type I diabetes in NOD.C3H congenic strains. Genomics 74:163eC171, 2001

    Ghosh S, Palmer SM, Rodrigues NR, Cordell HJ, Hearne CM, Cornall RJ, Prins J-B, McShane P, Lathrop GM, Peterson LB, Wicker LS, Todd JA: Polygenic control of autoimmune diabetes in nonobese diabetic mice. Nat Genet 4:404eC409, 1993

    Carnaud C, Gombert J, Donnars O, Garchon H, Herbelin A: Protection against diabetes and improved NK/NKT cell performance in NOD.NK1.1 mice congenic at the NK complex. J Immunol 166:2404eC2411, 2001

    Dallas-Pedretti A, McDuffie M, Haskins K: A diabetes-associated T-cell autoantigen maps to a telomeric locus on mouse chromosome 6. Proc Natl Acad Sci U S A 92:1386eC1390, 1995

    Pauza ME, Dobbs CM, He J, Patterson T, Wagner S, Anobile BS, Bradley BJ, Lo D, Haskins K: T-cell receptor transgenic response to an endogenous polymorphic autoantigen determines susceptibility to diabetes. Diabetes 53:978eC988, 2004

    Leijon K, Hammarstrom B, Holmberg D: Non-obese diabetic (NOD) mice display enhanced immune responses and prolonged survival of lymphoid cells. Int Immunol 6:339eC345, 1994

    Penha-Goncalves C, Leijon K, Persson L, Holmberg D: Type 1 diabetes and the control of dexamethazone-induced apoptosis in mice maps to the same region on chromosome 6. Genomics 28:398eC404, 1995

    Bergman ML, Duarte N, Campino S, Lundholm M, Motta V, Lejon K, Penha-Goncalves C, Holmberg D: Diabetes protection and restoration of thymocyte apoptosis in NOD Idd6 congenic strains. Diabetes 52:1677eC1682, 2003

    Bergman ML, Penha-Goncalves C, Lejon K, Holmberg D: Low rate of proliferation in immature thymocytes of the non-obese diabetic mouse maps to the Idd6 diabetes susceptibility region. Diabetologia 44:1054eC1061, 2001

    Ghiringhelli F, Larmonier N, Schmitt E, Parcellier A, Cathelin D, Garrido C, Chauffert B, Solary E, Bonnotte B, Martin F: CD4+CD25+ regulatory T cells suppress tumor immunity but are sensitive to cyclophosphamide which allows immunotherapy of established tumors to be curative. Eur J Immunol 34:336eC344, 2004

    Ablamunits V, Quintana F, Reshef T, Elias D, Cohen IR, Podolin PL, Denny P, Armitage N, Lord CJ, Hill NJ, Levy ER, Peterson LB, Todd JA, Wicker LS, Lyons PA: Acceleration of autoimmune diabetes by cyclophosphamide is associated with an enhanced IFN-gamma secretion pathway. J Autoimmun 13:383eC392, 1999

    Faveeuw C, Gagnerault MC, Lepault F: Isolation of leukocytes infiltrating the islets of Langerhans of diabetes-prone mice for flow cytometric analysis. J Immunol Methods 187:163eC169, 1995

    Yu L, Robles DT, Abiru N, Kaur P, Rewers M, Kelemen K, Eisenbarth GS: Early expression of antiinsulin autoantibodies of humans and the NOD mouse: evidence for early determination of subsequent diabetes. Proc Natl Acad Sci U S A 97:1701eC1706, 2000

    Thebault-Baumont K, Dubois-Laforgue D, Krief P, Briand JP, Halbout P, Vallon-Geoffroy K, Morin J, Laloux V, Lehuen A, Carel JC, Jami J, Muller S, Boitard C: Acceleration of type 1 diabetes mellitus in proinsulin 2-deficient NOD mice. J Clin Invest 111:851eC857, 2003

    Lepault F, Gagnerault MC, Faveeuw C, Bazin H, Boitard C: Lack of L-selectin expression by cells transferring diabetes in NOD mice: insights into the mechanisms involved in diabetes prevention by Mel-14 antibody treatment. Eur J Immunol 25:1502eC1507, 1995

    Schuler W, Weiler IJ, Schuler A, Phillips RA, Rosenberg N, Mak TW, Kearney JF, Perry RP, Bosma MJ, Asano M, Itoh M, Toda M: Rearrangement of antigen receptor genes is defective in mice with severe combined immune deficiency. Cell 46:963eC972, 1986

    Lepault F, Gagnerault MC: L-selectin(-/lo) and diabetogenic T cells are similarly distributed in pre-diabetic and diabetic nonobese diabetic mice. Lab Invest 78:551eC558, 1998

    Gregori S, Giarratana N, Smiroldo S, Adorini L: Dynamics of pathogenic and suppressor T cells in autoimmune diabetes development. J Immunol 171:4040eC4047, 2003

    Rohane PW, Shimada A, Kim DT, Edwards CT, Charlton B, Shultz LD, Fathman CG: Islet-infiltrating lymphocytes from pre-diabetic NOD mice rapidly transfer diabetes to NOD-scid/scid mice. Diabetes 44:550eC554, 1995

    Boitard C, Yasunami R, Dardenne M, Bach JF: T cell-mediated inhibition of the transfer of autoimmune diabetes in NOD mice. J Exp Med 169:1669eC1680, 1989

    Lepault F, Gagnerault MC: Characterization of peripheral regulatory CD4+ T cells that prevent diabetes onset in nonobese diabetic mice. J Immunol 164:240eC247, 2000

    Szanya V, Ermann J, Taylor C, Holness C, Fathman CG: The subpopulation of CD4+CD25+ splenocytes that delays adoptive transfer of diabetes expresses L-selectin and high levels of CCR7. J Immunol 169:2461eC2465, 2002

    Salomon B, Lenschow DJ, Rhee L, Ashourian N, Singh B, Sharpe A, Bluestone JA, Ho SC, Sattar H, Gray G, Nabavi N, Herold KC: B7/CD28 costimulation is essential for the homeostasis of the CD4+CD25+ immunoregulatory T cells that control autoimmune diabetes. Immunity 12:431eC440, 2000

    Myrick C, DiGuisto R, DeWolfe J, Bowen E, Kappler J, Marrack P, Wakeland EK: Linkage analysis of variations in CD 4:CD8 T cell subsets between C57BL/6 and DBA/2. Genes Immun 3:144eC150, 2002

    Stephens LA, Mason D, Hori S, Nomura T, Sakaguchi S: CD25 is a marker for CD4+ thymocytes that prevent autoimmune diabetes in rats, but peripheral T cells with this function are found in both CD25+ and CD25eC subpopulations. J Immunol 165:3105eC3110, 2000

    Seddon B, Saoudi A, Nicholson M, Mason D, Hori S, Nomura T, Sakaguchi S: CD4+CD8eC thymocytes that express L-selectin protect rats from diabetes upon adoptive transfer. Eur J Immunol 26:2702eC2708, 1996

    Sempe P, Richard MF, Bach JF, Boitard C: Evidence of CD4+ regulatory T cells in the non-obese diabetic male mouse. Diabetologia 37:337eC343, 1994

    Hoglund P, Mintern J, Waltzinger C, Heath W, Benoist C, Mathis D: Initiation of autoimmune diabetes by developmentally regulated presentation of islet cell antigens in the pancreatic lymph nodes. J Exp Med 189:331eC339, 1999

    Gagnerault MC, Luan JJ, Lotton C, Lepault F: Pancreatic lymph nodes are required for priming of beta cell reactive T cells in NOD mice. J Exp Med 196:369eC377, 2002

    Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M: Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25): breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol 155:1151eC1164, 1995

    Asano M, Toda M, Sakaguchi N, Sakaguchi S, Hori S, Nomura T: Autoimmune disease as a consequence of developmental abnormality of a T cell subpopulation. J Exp Med 184:387eC396, 1996

    Gershon RK, Kondo K: Cell interactions in the induction of tolerance: the role of thymic lymphocytes. Immunology 18:723eC737, 1970

    Hall BM, Pearce NW, Gurley KE, Dorsch SE: Specific unresponsiveness in rats with prolonged cardiac allograft survival after treatment with cyclosporine. III. Further characterization of the CD4+ suppressor cell and its mechanisms of action. J Exp Med 171:141eC157, 1990

    Bach JF: Regulatory T cells under scrutiny. Nat Rev Immunol 3:189eC198, 2003

    Pop SM, Wong CP, Culton DA, Clarke SH, Tisch R: Single cell analysis shows decreasing FoxP3 and TGF1 coexpressing CD4+CD25+ regulatory T cells during autoimmune diabetes. J Exp Med 201:1333eC1346, 2005

    Kono DH, Burlingame RW, Owens DG, Kuramochi A, Balderas RS, Balomenos D, Theofilopoulos AN: Lupus susceptibility loci in New Zealand mice. Proc Natl Acad Sci U S A 91:10168eC10172, 1994

    Zhang Z, Futamura M, Vikis HG, Wang M, Li J, Wang Y, Guan KL, You M: Positional cloning of the major quantitative trait locus underlying lung tumor susceptibility in mice. Proc Natl Acad Sci U S A 100:12642eC12647, 2003

    Maria DA, Manenti G, Galbiati F, Ribeiro OG, Cabrera WH, Barrera RG, Pettinicchio A, De Franco M, Starobinas N, Siqueira M, Dragani TA, Ibanez OM: Pulmonary adenoma susceptibility 1 (Pas1) locus affects inflammatory response. Oncogene 22:426eC432, 2003

    Manenti G, Peissel B, Gariboldi M, Falvella FS, Zaffaroni D, Allaria B, Pazzaglia S, Rebessi S, Covelli V, Saran A, Dragani TA: A cancer modifier role for parathyroid hormone-related protein. Oncogene 19:5324eC5328, 2000

    Khattri R, Cox T, Yasayko SA, Ramsdell F: An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nat Immunol 4:337eC342, 2003

    Fontenot JD, Gavin MA, Rudensky AY, Hori S, Nomura T, Sakaguchi S: Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 4:330eC336, 2003

    Hori S, Nomura T, Sakaguchi S: Control of regulatory T cell development by the transcription factor Foxp3. Science 299:1057eC1061, 2003

    Nakamura K, Kitani A, Fuss I, Pedersen A, Harada N, Nawata H, Strober W: TGF-beta1 plays an important role in the mechanism of CD4+CD25+ regulatory T cell activity in both humans and mice. J Immunol 172:834eC842, 2004(Ute Christine Rogner, Fra)