当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 糖尿病学杂志 > 2005年 > 第5期 > 正文
编号:11256502
Autoimmune Diabetes Onset Results From Qualitative Rather Than Quantitative Age-Dependent Changes in Pathogenic T-Cells
     1 Institut National de la Santee et de la Recherche Meedicale (INSERM) U580, Paris, France

    2 Facultee de Medicine, Renee Descartes Paris 5, Paris, France

    3 Sir William Dunn School of Pathology, Oxford, U.K

    ABSTRACT

    Diabetogenic T-cells can be detected in pre-diabetic nonobese diabetic (NOD) mice after transfer in NOD-SCID recipients. Here we demonstrate that 6-week-old pre-diabetic NOD mice, >2 months before disease onset, already harbor pathogenic T-cells in equal numbers to overtly diabetic animals. The delay in diabetes appearance is explained by the presence of regulatory CD4+CD25+ T-cells that control diabetogenic effectors and that are, in our hands, transforming growth factor (TGF)-eCdependent. Our present results suggest, however, that diabetes onset is only partly explained by a decline in this regulatory T-cell activity. Another major factor appears to be the progressive resistance of diabetogenic cells to TGF-eCdependent mediated inhibition. We propose that progression to overt disease correlates with the pathogenic T-cell’s escape from TGF-eCdependent T-celleCmediated regulation.

    Insulin-dependent diabetes (type 1 diabetes) is a genetically controlled autoimmune disease caused by selective destruction of insulin-secreting -cells by pathogenic T-cells (1). In nonobese diabetic (NOD) mice, a spontaneous model of type 1 diabetes, pathogenic T-cells are easily assessed by adoptive transfer into syngeneic immunodeficient recipients (1eC5). In NOD mice, rupture of self-tolerance is first evidenced at 3eC4 weeks of age when the initial wave of mononuclear cells infiltrates pancreatic islets that is 3eC4 months before the first signs of overt disease, as assessed by glycosuria and hyperglycemia (1,6,7). During this quite long phase (i.e., pre-diabetes), insulitis progresses until 12eC14 weeks of age as a "benign" process that is not associated with massive -cell destruction (1). Although major progress has been made in our knowledge on the candidate autoantigens (8eC10) and the forces driving the emergence of diabetogenic T-cells (1,11eC17), there are still uncertainties about the mechanisms underlying the progression to irreversible -cell destruction. There is compelling evidence to show that regulatory T-cells mediating transferable tolerance actively control diabetogenic effectors (18eC24). However, is this just a game of numbers Does diabetes onset exclusively result from either the decrease in regulatory T-cells over time or from the progressive expansion of -celleCspecific T-cell effectors overriding the control afforded by regulatory T-cells

    Our current results point to an alternative possibility, which is that there are key age-dependent differences in effector T-cells that are qualitative rather than quantitative and render them progressively insensitive to T-celleCmediated regulation. This insensitivity involves progressive unresponsiveness of effector T-cells to the immunoregulatory cytokine transforming growth factor (TGF)-.

    RESEARCH DESIGN AND METHODS

    NOD mice (Kd, I-Ag7, Db) and NOD-SCID mice were bred in our animal facility under specific pathogen-free conditions. Glycosuria and glycemia were monitored using colorimetric strips (Glukotest and Hemoglukotest; Boehringer-Mannheim).

    Fluorescence-activated cell sorting analysis.

    Lymphocytes were stained for surface markers and analyzed by flow cytometry. Antibodies to TGF- (2G.7) were purified and fluoresceinated in our laboratory. AntieCglucocorticoid-induced tumor necrosis factor receptor (GITR) antibodies were kindly provided by Dr. Cobbold (Sir William Dunn School of Pathology, Oxford, U.K.). CD62L, CD25, CD4, and CD103 (E integrin subunit) antibodies were obtained from PharMingen-BD (San Diego, CA).

    Cell preparations.

    Splenocytes were isolated from 4- or 6-week-old or diabetic NOD mice. Splenocytes were then purified on the basis of CD62L, CD25, or CD4 expression using magnetic bead cell sorting (Miltenyi Biotech, Bergisch-Gladbach, Germany). Purity of the sorted cells was 90eC97%, and recovery ranged from 50 to 70%.

    Adoptive cell transfers.

    Recipients were adult 6-week-old NOD-SCID mice. Animals were injected intravenously with either a single cell population or, in the case of cotransfer experiments, a mixture of two distinct populations. The precise cell numbers used varied depending on the experiments and are detailed in the RESULTS section.

    In vitro proliferation assays.

    Cells were cultured in complete RPMI 1640. CD4+CD25+ and CD4+CD25eC T-cells were seeded in 96-well microplates (2 x 104 cells/well) and stimulated with soluble anti-CD3 antibody (2.5 e/ml 145-2C11; provided by J.A. Bluestone) in the presence of mitomycin-treated antigen-presenting cells (APCs). Neutralizing antibodies to TGF- were added to the cocultures (10 or 50 e/ml 2G.7). For other experiments, CD4+CD25eC and CD4+CD25eCCD62LeC T-cells were incubated with APCs, anti-CD3 antibody, and recombinant active TGF- (0eC4,000 pg/ml; R&D Systems, Abingdon, U.K.). For "criss-cross" cocultures, CD4+CD25+ T-cells from 6-week-old animals were coincubated with CD4+CD25eC T-cells isolated from diabetic NOD mice, and, inversely, CD4+CD25eC T-cells from 6-week-old animals were coincubated with CD4+CD25+ T-cells isolated from diabetic NOD mice. After 72 h at 37°C, cells were pulsed with [3H]thymidine (Amersham). Data were expressed as the percent inhibition, deduced as: percent inhibition = {1 eC[cpm (CD4+CD25eC plus CD4+CD25+)/cpm CD4+CD25eC]} x 100, where cpm is the counts per minute.

    Similar assays were performed in 24-well plates using transwells (Costar). 5,6-carboxyfluorescein diacetate-succinimidyl estereClabeled CD4+CD25eC T-cells from 6-week-old NOD mice (2 x 105 cells/well) were stimulated with anti-CD3 antibody and APCs. CD4+CD25+ T-cells (2 x 105 cells/well) were added in the transwell with APCs. After 4 days, cells were stained with CD4 antibodies, and the CD4+CD25eC T-cell proliferation was analyzed by fluorescence-activated cell sorting.

    Cytokine production.

    CD4+CD25+ and CD4+CD25eC T-cells from 6-week-old or diabetic mice were used and cultured as described in the proliferation assay. "Age-matched" or "crossed" cocultures were performed, and supernatants were recovered after 24, 48, and 72 h of culture. Interleukin (IL)-4, IL-10, and -interferon (IFN-) were measured by enzyme-linked immunosorbent assays, and TGF-1 was measured using a DuoSet kit (R&D Systems).

    Real-time quantitative RT-PCR.

    DNase IeCtreated total RNA from tissues was prepared using the SV Total RNA isolation system (Promega, Madison, WI). Reverse transcription was performed using a proStar kit with random hexamers (Stratagene, Cedar Creek, TX). Real-time quantification was performed using gene-specific fluorogenic probes and a Universal MasterMix kit (PE-Applied Biosystem). PCR and TaqMan analysis were performed using the ABI/PRISM 7700 sequence detector system (PE-Applied Biosystem, Warrington, U.K.). Multiplex PCRs were performed using appropriate gene-specific primers and VIC-labeled CD3 and FAM-labeled foxP3 probes. Standard curves of cDNAs from normal spleen were used to calibrate CT to amounts of foxP3 and to normalize cDNAs on each 96-well plate run. Normalized values for mRNA expression were calculated as (foxP3 mean)/(CD3 mean).

    Statistical analysis.

    The occurrence of diabetes was plotted using the Kaplan-Meier method, i.e., a nonparametric cumulative survival plot. The statistical comparison between the curves was performed using the log-rank (Mantel-Cox) test. In addition, results were analyzed using Student’s t test when needed.

    RESULTS

    Diabetogenic cells are present in large numbers long before onset of overt diabetes.

    Adoptive transfer of CD4+ and CD8+ spleen cells from diabetic NOD mice into immunodeficient syngeneic recipients, i.e., NOD-SCID mice, triggers diabetes (18,20). Using unseparated splenocytes, disease transfer was not observed after injection of cells from 4-week-old donors (data not shown). Diabetes transfer was, however, obtained on injection of cells from pre-diabetic donors aged 6 weeks, but later and at a lower rate when compared with that observed on transfer of total spleen cells from diabetic donors (Fig. 1A). This difference could be explained either by the progressive accumulation of diabetogenic T-cell effectors present in sufficient numbers only in overtly diabetic animals, or alternatively by the presence in pre-diabetic animals of subsets of regulatory T-cells mediating active tolerance whose number and/or functional capacity would decline with disease progression. To distinguish between these two hypotheses, transfer experiments were performed using splenocytes from NOD mice at different ages depleted of regulatory T-cells, i.e., expressing the -chain of the IL-2 receptor (CD25) or L-selectin (CD62L) (3,20,22eC24). Results showed that diabetes incidence was similar in recipients injected with equal numbers of CD25eCCD62LeC (e.g., enriched for effector cells) from 6-week-old NOD or diabetic mice (including same proportions of CD4+ and CD8+ T-cells); however, there was a slight delay in disease onset in recipients of effector cells from the younger mice (Fig. 1B). Such a pattern was comparable to that observed on transfer of total splenocytes from diabetic mice (Fig. 1A). In contrast, CD25eCCD62LeC cells from pre-diabetic 4-week-old NOD mice did not transfer diabetes.

    These data show that fully competent diabetogenic CD25eCCD62LeC cells appear at the periphery between 4 and 6 weeks of age. Moreover, diabetogenic cells from pre-diabetic 6-week-old mice and those from diabetic mice appear equally potent at transferring diabetes. Because spontaneous diabetes only appears 10 weeks later, these results stress the role of regulatory T-cells expressing CD25 and/or CD62L in controlling disease progression.

    In vivo sensitivity of diabetogenic effectors to T-celleCmediated regulation.

    We next explored whether the deficient suppression observed in mice with overt disease was caused by either decreased functional capacity of regulatory T-cells or diminished effector cell sensitivity to regulatory cells, or both. To this end, we performed criss-cross in vivo experiments to analyze the sensitivity of diabetogenic cells from young or old NOD mice to regulatory T-cells recovered from young or old NOD donors.

    In all of the experiments CD25eCCD62LeC spleen cells, depleted in CD25+ and CD62L+ subsets, were used as effector diabetogenic cells. Figure 2 shows that CD4+CD25+ T-cells (e.g., enriched for regulatory T-cells) from young NOD mice were less efficient at controlling diabetes transfer induced by effector cells from diabetic mice than that elicited by injection of effector cells from young pre-diabetic donors. This was observed using equal numbers of effector cells (CD25eCCD62eC cells) from two origins that, as mentioned above, were equally potent at transferring diabetes (Fig. 1B). Moreover, CD4+CD25+ T-cells from diabetic NOD mice significantly inhibited disease transfer elicited by effector cells from young NOD mice, albeit less efficiently than CD4+CD25+ T-cells from 6-week-old animals (Fig. 2). Thus, at the time of diabetes onset, the age-dependent reduction in the functional capacity of regulatory cells is only partial and parallels a significantly reduced sensitivity of diabetogenic effectors to T-celleCmediated regulation.

    Age-dependent decline in the in vitro sensitivity of CD4+CD25eC T-cells to CD4+CD25+ T-cells.

    To further investigate the cellular mechanisms mediating the resistance of effector cells to immunoregulation, we performed in vitro studies using the conventional noneCantigen-specific coculture model. CD4+CD25+ T-cells have the capacity to suppress the proliferation of CD4+CD25eC T-cells in coculture (25eC28). As detailed in Fig. 3A, a consistent age-dependent decrease in in vitro suppressive ability was observed in NOD mice. CD4+CD25+ T-cells from young 6-week-old pre-diabetic mice efficiently inhibited the proliferation of CD4+CD25eC T-cells from the same donors (Fig. 3A). In contrast to this, a significantly lower suppression was found using CD4+CD25+ cells from 8-week-old NOD mice cocultured with CD4+CD25eC T-cells from the same donors (Fig. 3A). Moreover, very low or no suppression (ranging from 0 to 10%) was found in 50% of cases when CD4+CD25+ and CD4+CD25eC cells were collected from the same overtly diabetic donors (Fig. 3A). These low suppression indexes were not explained by an insufficient ratio of suppressor to effector cells because no significant effect was observed when increasing the CD25+-to-CD25eC ratio (Fig. 3B).

    The in vitro functional capacity of CD4+CD25+ T-cells recovered from NOD H-2k and B6 H-2g7 mice (used as controls) at 6, 16, and 35 weeks of age was not significantly different from that found in young pre-diabetic NOD mice and did not vary with aging, suggesting a genetic dependency of the decline of the suppressive ability in the NOD strain (data not shown). In normal noneCautoimmune-prone mice, the CD4+CD25+-mediated suppressor activity was cell-to-cell contact dependent (25). Using 5,6-carboxyfluorescein diacetate-succinimidyl estereClabeledCD4+CD25eC T-cells, we observed that also in NOD mice the proportion of dividing cells significantly increased when CD4+CD25+ T-cells were physically separated from their targets using the transwell device (Fig. 3C).

    We next analyzed whether the deficient in vitro suppression observed in diabetic mice was caused not only by decreased functional capacity of regulatory T-cells but also by a diminished sensitivity of proliferating cells to immunoregulation. Criss-cross in vitro experiments were performed, using a similar strategy to that described above for the in vivo cotransfers. Figure 4 shows that CD4+CD25+ T-cells from diabetic NOD mice suppressed proliferation of CD4+CD25eC T-cells from young pre-diabetic mice, though less efficiently than CD4+CD25+ T-cells from young NOD mice. Conversely, CD4+CD25+ T-cells from young NOD mice were as ineffective as CD4+CD25+ T-cells from diabetic mice at suppressing the proliferation of CD4+CD25eC T-cells from overtly diabetic donors.

    We analyzed the expression of several regulatory T-cell candidate markers on the various T-cell subsets that showed a protective or diabetogenic ability. FoxP3 mRNA was detected in CD4+CD25+ T-cells but not in CD4+CD25eC T-cells (Fig. 5A). Surprisingly, CD4+CD25+ T-cells from diabetic animals expressed higher levels of foxP3 mRNA compared with those from 6-week-old NOD mice. The GITR was also exclusively expressed on CD4+CD25+ T-cells (Fig. 5B), with no significant difference between young and diabetic NOD mice. Finally, 18eC20% of CD4+CD25+ T-cells expressed CD103 (E subunit of the E7 integrin); similar levels and intensity were observed in young and diabetic NOD mice.

    Age-dependent sensitivity of effector cells to TGF-.

    TGF- has been shown to be involved in CD4+ T-celleCmediated regulation both in vivo and in vitro (25,29eC34). We examined the ability of neutralizing antieCTGF- antibodies to alter T-celleCmediated regulation in NOD mice. First, we observed that the in vitro suppression was abrogated in a dose-dependent manner on addition of an antieCTGF- antibody (2G.7; 49.1 ± 4.5 and 92.2 ± 2.1% reversal of suppression [mean ± SE] for 10 and 50 e/ml of antibody, respectively) (Fig. 3A). This is also in keeping with our data showing that in vivo, in the cotransfer model described above, administration of antieCTGF- antibodies reversed protection from disease transfer afforded by CD4+CD25+ T-cells from young NOD mice (data not shown).

    On in vitro stimulation with anti-CD3 antibody, CD4+CD25+ T-cells from young pre-diabetic NOD mice produced TGF- at levels significantly higher that those produced by the equivalent subset from diabetic mice (Table 1). In addition, membrane TGF- was spontaneously expressed, in the absence of any previous stimulation, on 13% of CD4+CD25+ T-cells independent of the age of the mouse (Fig. 5B). CD4+CD25eC T-cells expressed very low levels of membrane TGF- (2eC4%).

    IL-10 was produced in higher amounts by CD4+CD25+ T-cells from young animals, whereas the opposite pattern was observed for IFN- (Table 1). We also observed that CD4+CD25eC T-cells from diabetic animals produced fourfold higher levels of IFN- compared with those recovered from 6-week-old mice. Similarly, in coculture, higher TGF- concentrations were observed when CD4+CD25+ T-cells from young animals were present, whereas high IFN- production was observed when CD4+CD25eC T-cells from diabetic animals were present (Table 2).

    Given the central role of TGF-, it was important to study more directly its influence on effector cells. This was performed in vitro by adding increasing amounts of recombinant TGF- on anti-CD3 antibodyeCstimulated CD4+CD25eC and CD4+CD25eCCD62LeC T-cells from pre-diabetic and diabetic NOD mice. Importantly, for all T-cell subsets studied, strictly identical cell numbers were used. Proliferating CD4+CD25eC and CD4+CD25eCCD62LeC T-cells from 6-week-old NOD mice were significantly more sensitive to TGF-eCmediated inhibition than those from diabetic mice (Fig. 6). Ten to twenty times more recombinant TGF- was necessary to suppress proliferation of pathogenic cells recovered from diabetic animals compared with those recovered from young animals. Expression of TGF- receptors I and II was investigated by Western blot, and both were detected on CD4+CD25eCCD62LeC T-cells recovered from young NOD mice as well as diabetic animals (data not shown). In addition, the expression of TGF- receptor signaling molecules such as phosphorylated Smad2, also assessed by Western blot, did not appear to significantly differ in extracts from effector T-cells recovered from young NOD mice and diabetic mice (data not shown).

    DISCUSSION

    Direct evidence for loss of -celleCspecific self-tolerance as assessed by mononuclear cell infiltration of pancreatic islets is present very early in NOD mice (3 weeks of age), a long time before onset of overt disease (1,6,7). So far, it has been generally assumed that this is mainly caused by a "game of numbers" in which islet-specific pathogenic T-cells progressively accumulate, overriding T-celleCmediated regulation that controls pathogenic aggressors (7,18eC24). This is also compatible with the observation that NOD mice have a general failure in tolerance caused by defective negative selection in the thymus (35,36). However, this emergence of autoreactive T-cells in abnormally high numbers is not sufficient per se. In addition, there is compelling evidence to suggest that -cell autoantigen(s)eCdriven T-cell activation is a mandatory step for the emer-ence of the aggressive "diabetogenic" lymphocyte response (1,8eC10,12).

    Our current data are the first to demonstrate that, besides quantitative changes that might occur in autoantigen-specific T-cell populations as disease progresses, autoimmune diabetes also results from qualitative differences in diabetogenic T-cells. As assessed in adoptive transfer, pathogenic effectors are detectable in young pre-diabetic 6-week-old NOD mice. These diabetogenic T-cells can only be identified if purified on the basis of their lack of both CD25 and CD62L (3,4) to eliminate regulatory T-cells that effectively slow down diabetes transfer when total spleen cell populations are used.

    Our data also confirm the reported important role of regulatory CD4+CD25+ and CD62L+ T-cells in controlling pathogenic effectors (18,20eC23,37). Thus, B7eC/eC and CD28eC/eC NOD mice, which lack CD4+CD25+ T-cells, show accelerated disease onset (23). Infusion of wild-type syngeneic CD25+ T-cells back into CD28eC/eC NOD recipients prevents disease (23). Regulatory T-cells present in NOD mice share some similarities with those described in other models, notably the polyautoimmune syndrome induced by day 3 thymectomy in BALB/c mice (38,39) and diabetes observed after thymectomy and sublethal irradiation of PVG rats (30,40). They express high levels of the transcription factor foxP3 and other membrane molecules considered "candidate" markers for regulatory T-cells such as GITR (41,42), membrane TGF- (25,31), and CD103 (43). Moreover, in vitro they effectively suppress the anti-CD3 antibodyeCinduced proliferation of autologous CD4+CD25eC T-cells.

    Given this high number of diabetogenic cells in young pre-diabetic mice, we wanted to understand why it is only 2eC3 months later that overt irreversible disease develops. The first hypothesis was that regulatory T-cells progressively decrease in numbers and/or functional capacity until they become incapable of controlling pathogenic effectors. Results from in vivo cotransfer experiments mixing diabetogenic and regulatory T-cells from the same donors (either young pre-diabetic or overtly diabetic) show that good protection from disease transfer is obtained with regulatory T-cells from young, but not aged donors, which would favor this explanation. However, the situation is more complex. In criss-cross cotransfers it became apparent that regulatory T-cells from diabetic NOD mice still protected from disease transfer caused by CD25eCCD62LeC pathogenic cells from young donors. In addition, CD25eCCD62LeC diabetogenic T-cells from diabetic donors were insensitive to the protective effect of CD4+CD25+ T-cells from young NOD mice that appeared highly protective when cotransferred with CD25eCCD62LeC cells from young donors.

    In vitro, regulatory T-cells from young NOD mice efficiently inhibited anti-CD3 antibodyeCinduced proliferation of CD4+CD25eC T-cells from the same donors but were ineffective at suppressing CD4+CD25eC T-cells from diabetic mice. Moreover, regulatory T-cells from diabetic NOD mice did not efficiently suppress proliferation of CD4+CD25eC T-cells from diabetic mice, whereas they were operational (albeit less efficiently than regulatory T-cells from young donors) on CD4+CD25eC T-cells from young mice. Thus, in such a noneCantigen-specific system, results fully paralleled the data from in vivo criss-cross transfers discussed above.

    Therefore, although regulatory T-cells appeared less effective in diabetic than in young NOD mice, they were still functional at the time of overt diabetes. This is well in keeping with the observation that the proportions of regulatory T-cells (i.e., CD25+, CD62L+, and GITR+) did not decrease with aging. Concerning foxP3 expression, it was in fact higher in diabetic versus young NOD mice. Besides these qualitative interpretations of the results, quantitative arguments cannot be excluded. Although CD25+ T-cells are certainly enriched for cells exhibiting a regulatory phenotype, this does not imply that all CD25+ T-cells express regulatory functions. In diabetic mice CD4+CD25+ T-cells may in fact include activated nonregulatory T-cells.

    In our hands, the in vitro suppression assay was cell-to-cell contact dependent, and TGF-, both soluble and membrane-bound, appeared to play a central role, as shown by the dose-dependent effective reversal of suppression observed on addition of a neutralizing TGF- antibody. The relevance of TGF-, initially described in the mouse (25) and subsequently in humans (44), has been challenged by the observations showing that CD4+CD25+ T-cells from TGF-eCdeficient mice could mediate effective suppression (28). It is fair, however, to quote the various reports arguing for the role of TGF- in T-celleCmediated regulation, although it is still difficult to conclude on its action as a mediator of the regulation and/or as a factor promoting the differentiation of regulatory cells. Among those reports are the oral tolerance models (33,34) and the ones showing protection from autoimmune diabetes mediated by transgenic expression of TGF-1 in pancreatic islets (45), by TGF-eCproducing CD4+ T-cell clones (46,47), or by islet-infiltrating CD4+CD25+ T-cells (31). Neutralizing TGF- antibodies have also been shown to reverse the ability of distinct subsets of CD4+ regulatory T-cells to protect from autoimmune or immune-mediated diseases (29,30).

    We further investigated whether the age-dependent decrease in sensitivity of diabetogenic effectors to immunoregulation involved TGF-. Indeed, CD4+CD25eC and CD4+CD25eCCD62LeC T-cells from young NOD mice were more sensitive than those from diabetic animals to the effect of recombinant TGF- (i.e., they required less TGF- to be negatively regulated). Mechanisms underlying this defect must be elucidated. Preliminary Western blot analysis revealed the presence of TGF- receptors I and II on the diabetogenic cells at a similar level, irrespective of the age of the donors. However, it has to be determined whether this upregulated resistance to TGF- in overtly diabetic mice relies on variations in the number of TGF- receptors and/or in their signaling capacity, including variations in the level of expression of molecules such as Smad7. Correlating with this enhanced resistance to regulation, CD4+CD25eC T-cells from older mice exhibited an accentuated Th1 phenotype. Thus, higher IFN-eCtoeCIL-10 and IFN-eCtoeCIL-4 ratios were observed in diabetic versus young mice.

    To conclude, our results indicate that the buildup of a large pool of diabetogenic T-cells in NOD mice occurs very early and is not sufficient to promote disease onset, thus stressing the role of regulatory T-cells in diabetes control (3,19,22eC24,31). In addition to this, we provide new information that disease onset is associated with loss of sensitivity of effector cells to TGF-eCdependent T-celleCmediated regulation. This concept is reminiscent of different in vitro and in vivo previous data (37,48,49). In particular, in vivo data in lupus-prone mice (NZB/NZW) showed that increased antierythrocyte autoantibody production was insensitive to regulatory T-cells, which were detectable in these animals (48). More recently, in autoimmune-prone Cbl-beC/eC mice that express CD28-independent T-cell hyper-reactivity (50), effector CD4+CD25eC T-cells were shown to be resistant to both TGF- and CD4+CD25+ regulatory T-cells, which were present and functional in these mice (49).

    The question remains of the mechanisms underlying this resistance. One possibility is that at a certain point during the disease process, pathogenic T-cells express an autoantigen-dependent increase in their activation or avidity. This interpretation does not explain, however, the results obtained in our in vitro cocultures that were based on a noneCantigen-specific system. Another possibility is an age-dependent change in the sensitivity of effector T-cells to regulatory cytokines, namely TGF- in autoimmune-prone mice. Independent of interpretation, these data open interesting new therapeutic perspectives aimed at the selective elimination of the subset of TGF-eCresistant effector cells or at their pharmacological modulation, using agents affecting TGF-eCspecific receptors and/or their signal transduction pathways.

    ACKNOWLEDGMENTS

    This work was supported by grants from INSERM (Institut National de la Santee et de la Recherche Meedicale), FRM (Fondation pour la Recherche Meedicale), and the Juvenile Diabetes Research Foundation.

    The authors are indebted to F. Valette and M. Garcia for managing the animal facility, to M. Netter for the iconography, to Jeerme Meegret for assistance with fluorescence-activated cell sorting, to Anne Esling for technical assistance, and to Mary de la Torre for editing of the manuscript. We also thank W. Paul and A. O’Garra for providing monoclonal antibodies used in this study as well as J.A. Bluestone for providing monoclonal antibodies and for very helpful critical discussion.

    APC, antigen-presenting cell; GITR, glucocorticoid-induced tumor necrosis factor receptor; IFN-, -interferon; IL, interleukin; TGF, transforming growth factor

    REFERENCES

    Bach JF: Insulin-dependent diabetes mellitus as an autoimmune disease. Endocrine Rev15 :516 eC542,1994

    Wicker LS, Miller BJ, Mullen Y: Transfer of autoimmune diabetes mellitus with splenocytes from nonobese diabetic (NOD) mice. Diabetes35 :855 eC860,1986

    Alyanakian MA, You S, Damotte D, Gouarin C, Esling A, Garcia C, Havouis S, Chatenoud L, Bach JF: Diversity of regulatory CD4+T cells controlling distinct organ-specific autoimmune diseases. Proc Natl Acad Sci U S A100 :15806 eC15811,2003

    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 Immunol25 :1502 eC1507,1995

    Yagi H, Matsumoto M, Kunimoto K, Kawaguchi J, Makino S, Harada M: Analysis of the roles of CD4+ and CD8+ T cells in autoimmune diabetes of NOD mice using transfer to NOD athymic nude mice. Eur J Immunol22 :2387 eC2393,1992

    Makino S, Kunimoto K, Muraoka Y, Mizushima Y, Katagiri K, Tochino Y: Breeding of a non-obese, diabetic strain of mice. Jikken Dobutsu29 :1 eC13,1980

    Katz JD, Wang B, Haskins K, Benoist C, Mathis D: Following a diabetogenic T cell from genesis through pathogenesis. Cell74 :1089 eC1100,1993

    Kaufman DL, Clare-Salzler M, Tian J, Forsthuber T, Ting GSP, Robinson P, Atkinson MA, Sercarz EE, Tobin AJ, Lehmann PV: Spontaneous loss of T-cell tolerance to glutamic acid decarboxylase in murine insulin-dependent diabetes. Nature366 :69 eC72,1993

    Tisch R, Yang XD, Singer SM, Liblau RS, Fugger L, Mcdevitt HO: Immune response to glutamic acid decarboxylase correlates with insulitis in non-obese diabetic mice. Nature366 :72 eC75,1993

    Wegmann DR, Norbury-Glaser M, Daniel D: Insulin-specific T cells are a predominant component of islet infiltrates in pre-diabetic NOD mice. Eur J Immunol24 :1853 eC1857,1994

    Hglund P, Mintern J, Waltzinger C, Heath W, Benoist C, Mathis D: Initiation of autoimmune diabetes by developmentally regulated presentation of islet cell antigen in the pancreatic lymph nodes. J Exp Med189 :331 eC339,1999

    Larger E, Becourt C, Bach JF, Boitard C: Pancreatic islet beta-cells drive T-cell immune responses in the nonobese diabetic mouse model. J Exp Med181 :1635 eC1642,1995

    Wong S, Guerder S, Visintin I, Reich EP, Swenson KE, Flavell RA, Janeway CA: Expression of the co-stimulator molecule B7eC1 in pancreatic beta-cells accelerates diabetes in the NOD mouse. Diabetes44 :326 eC329,1995

    Green EA, Flavell RA: Tumor necrosis factor-alpha and the progression of diabetes in non-obese diabetic mice. Immunol Rev169 :11 eC22,1999

    Trembleau S, Penna G, Bosi E, Mortara A, Gately MK, Adorini L: Interleukin 12 administration induces T helper type 1 cells and accelerates autoimmune diabetes in NOD mice. J Exp Med181 :817 eC821,1995

    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.Ig mu null mice. J Exp Med184 :2049 eC2053,1996

    Horwitz MS, Bradley LM, Harbertson J, Krahl T, Lee J, Sarvetnick N: Diabetes induced by Coxsackie virus: initiation by bystander damage and not molecular mimicry. Nat Med4 :781 eC785,1998

    Boitard C, Yasunami R, Dardenne M, Bach JF: T cell-mediated inhibition of the transfer of autoimmune diabetes in NOD mice. J Exp Med169 :1669 eC1680,1989

    Sempe P, Richard MF, Bach JF, Boitard C: Evidence of CD4+ regulatory T cells in the non-obese diabetic male mouse. Diabetologia37 :337 eC343,1994

    Herbelin A, Gombert JM, Lepault F, Bach JF, Chatenoud L: Mature mainstream TCR alpha beta(+)CD4(+) thymocytes expressing L-selectin mediate "active tolerance" in the nonobese diabetic mouse. J Immunol161 :2620 eC2628,1998

    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 Immunol169 :2461 eC2465,2002

    Lepault F, Gagnerault MC: Characterization of peripheral regulatory CD4(+) T cells that prevent diabetes onset in nonobese diabetic mice. J Immunol164 :240 eC247,2000

    Salomon B, Lenschow DJ, Rhee L, Ashourian N, Singh B, Sharpe A, Bluestone JA: B7/CD28 costimulation is essential for the homeostasis of the CD4+CD25+ immunoregulatory T cells that control autoimmune diabetes. Immunity12 :431 eC440,2000

    Chatenoud L, Salomon B, Bluestone JA: Suppressor T cells—they’re back and critical for regulation of autoimmunity! Immunol Rev182 :149 eC163,2001

    Nakamura K, Kitani A, Strober W: Cell contact-dependent immunosuppression by CD4(+)CD25(+) regulatory T cells is mediated by cell surface-bound transforming growth factor beta. J Exp Med194 :629 eC644,2001

    Thornton AM, Shevach EM: Suppressor effector function of CD4+CD25+ immunoregulatory T cells is antigen nonspecific. J Immunol164 :183 eC190,2000

    Takahashi T, Kuniyasu Y, Toda M, Sakaguchi N, Itoh M, Iwata M, Shimizu J, Sakaguchi S: Immunologic self-tolerance maintained by CD25+CD4+ naturally anergic and suppressive T cells: induction of autoimmune disease by breaking their anergic/suppressive state. Int Immunol10 :1969 eC1980,1998

    Piccirillo CA, Letterio JJ, Thornton AM, Mchugh RS, Mamura M, Mizuhara H, Shevach EM: CD4(+)CD25(+) regulatory T cells can mediate suppressor function in the absence of transforming growth factor beta1 production and responsiveness. J Exp Med196 :237 eC246,2002

    Powrie F, Carlino J, Leach MW, Mauze S, Coffman RL: A critical role for transforming growth factor-beta but not interleukin 4 in the suppression of T helper type 1-mediated colitis by CD45RB(low) CD4+ T cells. J Exp Med183 :2669 eC2674,1996

    Seddon B, Mason D: Regulatory T cells in the control of autoimmunity: the essential role of transforming growth factor beta and interleukin 4 in the revention of autoimmune thyroiditis in rats by peripheral CD4(+)CD45RC-cells and CD4(+)CD8(-) thymocytes. J Exp Med189 :279 eC288,1999

    Green EA, Gorelik L, McGregor CM, Tran EH, Flavell RA: CD4+CD25+ T regulatory cells control anti-islet CD8+ T cells through TGF-beta-TGF-beta receptor interactions in type 1 diabetes. Proc Natl Acad Sci U S A100 :10878 eC10883,2003

    Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos N, McGrady G, Wahl SM: Conversion of peripheral CD4+CD25eCnaive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J Exp Med198 :1875 eC1886,2003

    Miller A, Lider O, Roberts AB, Sporn MB, Weiner HL: Suppressor T cells generated by oral tolerization to myelin basic protein suppress both in vitro and in vivo immune responses by the release of transforming growth factor beta after antigen-specific triggering. Proc Natl Acad Sci U S A89 :421 eC425,1992

    Hancock WW, Polanski M, Zhang J, Blogg N, Weiner HL: Suppression of insulitis in non-obese diabetic (NOD) mice by oral insulin administration is associated with selective expression of interleukin-4 and -10, transforming growth factor-beta, and prostaglandin-E. Am J Pathol147 :1193 eC1199,1995

    Kishimoto H, Sprent J: A defect in central tolerance in NOD mice. Nat Immunol2 :1025 eC1031,2001

    Lesage S, Hartley SB, Akkaraju S, Wilson J, Townsend M, Goodnow CC: Failure to censor forbidden clones of CD4 T cells in autoimmune diabetes. J Exp Med196 :1175 eC1188,2002

    Gregori S, Giarratana N, Smiroldo S, Adorini L: Dynamics of pathogenic and suppressor T cells in autoimmune diabetes development. J Immunol171 :4040 eC4047,2003

    Asano M, Toda M, Sakaguchi N, Sakaguchi S: Autoimmune disease as a consequence of developmental abnormality of a T cell subpopulation. J Exp Med184 :387 eC396,1996

    Suri-payer E, Amar AZ, Thornton AM, Shevach EM: CD4+CD25+ T cells inhibit both the induction and effector function of autoreactive T cells and represent a unique lineage of immunoregulatory cells. J Immunol160 :1212 eC1218,1998

    Stephens LA, Mason D: 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 CD25(-) subpopulations. J Immunol165 :3105 eC3110,2000

    Mchugh RS, Whitters MJ, Piccirillo CA, Young DA, Shevach EM, Collins M, Byrne MC: CD4(+)CD25(+) immunoregulatory T cells: gene expression analysis reveals a functional role for the glucocorticoid-induced TNF receptor. Immunity16 :311 eC323,2002

    Tone M, Tone Y, Adams E, Yates SF, Frewin MR, Cobbold SP, Waldmann H: Mouse glucocorticoid-induced tumor necrosis factor receptor ligand is costimulatory for T cells. Proc Natl Acad Sci U S A100 :15059 eC15064,2003

    Lehmann J, Huehn J, de la Rosa M, Maszyna F, Kretschmer U, Krenn V, Brunner M, Scheffold A, Hamann A: Expression of the integrin alpha Ebeta 7 identifies unique subsets of CD25+ as well as CD25-regulatory T cells. Proc Natl Acad Sci U S A99 :13031 eC13036,2002

    Levings MK, Sangregorio R, Sartirana C, Moschin AL, Battaglia M, Orban PC, Roncarolo MG: Human CD25+CD4+ T suppressor cell clones produce transforming growth factor beta, but not interleukin 10, and are distinct from type 1 T regulatory cells. J Exp Med196 :1335 eC1346,2002

    King C, Davies J, Mueller R, Lee MS, Krahl T, Yeung B, O’connor E, Sarvetnick N: TGF-beta1 alters APC preference, polarizing islet antigen responses toward a Th2 phenotype. Immunity8 :601 eC613,1998

    Han HS, Jun HS, Utsugi T, Yoon JW: Molecular role of TGF-beta, secreted from a new type of CD4+ suppressor T cell, NY4.2, in the prevention of autoimmune IDDM in NOD mice. J Autoimmun10 :299 eC307,1997

    Zekzer D, Wong FS, Wen L, Altieri M, Gurlo T, Von Grafenstein H, Sherwin RS: Inhibition of diabetes by an insulin-reactive CD4 T-cell clone in the nonobese diabetic mouse. Diabetes46 :1124 eC1132,1997

    Blanchard D, Bach MA: Thymic function in NZB mice. III. Impairment of the activity of specific suppressor cells involved in the regulation of antibody production against sheep red blood cells. Clin Exp Immunol42 :1 eC9,1980

    Wohlfert EA, Callahan MK, Clark RB: Resistance to CD4+CD25+ regulatory T cells and TGF-beta in Cbl-b-/- mice. J Immunol173 :1059 eC1065,2004

    Chiang YJ, Kole HK, Brown K, Naramura M, Fukuhara S, Hu RJ, Jang IK, Gutkind JS, Shevach E, Gu H: Cbl-b regulates the CD28 dependence of T-cell activation. Nature403 :216 eC220,2000(Sylvaine You, Meeriam Bel)