Dendritic cells can facilitate allograft survival and prevent autoimmunity via direct and indirect cell-mediated mechanisms. Recent studies demonstrate that immunoregulatory dendritic cells (iDCs) confer immune hyporesponsiveness in part through CD4+ CD25+ T regulatory cells (Tregs). Herein, we provide evidence to support the hypothesis that dendritic cells derived from NOD mice and engineered ex vivo to exhibit suppressed expression of the CD40, CD80, and CD86 costimulatory molecules motivate an increase in the prevalence of regulatory CD4+ CD25+ T-cells via interleukin (IL)-7. Unlike control dendritic cells, these dendritic cells expressed significant levels of IL-7. Exogenous addition of IL-7 to NOD T-cells did not promote expansion or proliferation, but instead selectively maintained the number of CD4+ CD25+ T-cells by inhibiting activation of apoptosis in these cells. In vitro, IL-7 receptor α-chain (IL-7Rα) was expressed at significantly higher levels on CD4+ CD25+ T-cells compared with CD4+ CD25 T-cells irrespective of resting or stimulated state. In vivo, CD4+ CD25+ T-cells obtained from NOD-scid mice reconstituted with ex vivo engineered iDCs and NOD splenocytes expressed significantly higher levels of IL-7Rα compared with levels in the CD4+ CD25 subset, especially in diabetes-suppressive dendritic cell–administered NOD-scid recipients. Taken together, our data suggest a novel mechanism by which iDCs delay autoimmunity through the CD4+ CD25+ Treg pathway and suggest IL-7 as a survival factor for these putative Tregs, which express the α-chain of its receptor at considerably higher levels than CD4+ CD25 T-cells.

Dendritic cells have been used in a variety of embodiments to promote allograft survival and suppress autoimmunity (14). A number of mechanisms appear to be active in conferring these properties in vivo in these “immunoregulatory” dendritic cells (iDCs; diabetes-delaying dendritic cell embodiments). These mechanisms include attenuated coactivation through molecules of the B7 family, decreased production of proinflammatory interleukins like interleukin (IL)-12, Fas ligand–induced apoptosis of T-cells, and the induction of indoleamine-2,3-dioxygenase, which prevents T-cell proliferation through tryptophan catabolism (115). More recently, iDCs have been shown to achieve immunosuppression through the enhancement of regulatory T-cell networks and, in particular, the CD4+ CD25+ T-cell subtype (1625).

Our recent studies showed that dendritic cells derived from the bone marrow progenitors of the NOD mice engineered to maintain low levels of CD40, CD80, and CD86 costimulatory molecules conferred a delay in type 1 diabetes incidence to syngeneic recipients (25). These dendritic cells promoted an increased steady-state number of CD4+ CD25+ T-cells when injected into pre-diabetic NOD mice without skewing the prevalence of T-cells producing TH2-type cytokines or toward a TR1/TR1-like phenotype. Furthermore, we observed no evidence that indoleamine-2,3-dioxygenase expression or activity was increased in our embodiment of diabetes-suppressive dendritic cells (M.W., J.M., N.G., unpublished observations). These data compelled us to determine alternative or complementary mechanisms by which the dendritic cells promoted increased numbers of CD4+ CD25+ putative regulatory T-cells. We hypothesized that dendritic cell–derived homeostatic maintenance regulatory proteins could partly explain how iDCs promoted an increased prevalence of putative T regulatory cells (Tregs) of CD4+ CD25+ phenotype. One such homeostatic regulator is the IL-7 cytokine.

IL-7 is a potent cytokine that plays a pivotal role in T-cell homeostasis (2630). IL-7 modulates mature T-cell function by acting as a surrogate costimulatory factor (2630), by enhancing T-cell proliferation and cytokine production, especially under suboptimal T-cell triggering (31). Although a proportion of these effects is IL-2 dependent, some are IL-2 independent (32,33). The large body of evidence to date demonstrates that IL-7 induces mainly TH1-type responses in upregulating IL-2 and interferon-γ production and acts in synergy with IL-12 on T-cells (31,34,35). However, the critical function served by IL-7 as a homeostatic regulator is in inhibiting cell death through apoptosis largely through the bcl family of antiapoptotic molecules (3641).

Recent data suggest that Tregs can be generated in the absence of IL-2 and that a complementary regulator could exist to maintain their number (42,43). IL-2 signaling passes largely through two main pathways. One leads to the activation of the Akt kinase cascade and the upregulation of antiapoptotic proteins and activity (44,45). The other passes through the activation of the signal transducer and activation of transcription (STAT)5 transcription factor and results in T-cell proliferation and differentiation (4447). A subset of mice deficient in STAT5A/5B showed autoimmune phenomena similar to those lacking IL-2 or its receptor and correlated with decreased numbers of CD4+ CD25+ Tregs (48). Tregs in these doubly deficient mice underwent apoptosis at an accelerated rate (48). Farrar and colleagues (49,50) have since proposed that the activation of an IL-7–sensitive transcription factor (STAT5) is important in the development or survival of CD4+ C25+ Tregs. Using a transgenic mouse expressing a constitutively activated STAT5 in B- and T-cell compartments, these studies showed that CD8+ T-cell numbers were dramatically increased with a cell surface phenotype identical to that of cells undergoing homeostatic proliferation or with a memory-like state (49). Additionally, they showed that these transgenic mice were no longer dependent on IL-15 or IL-7 to undergo homeostatic proliferation. Moreover, they demonstrated an increase in prevalence of CD4+ CD25+ Tregs expressing the Foxp3 transcription factor. Finally, these Tregs were capable of suppressing the proliferation of CD4+ CD25 wild-type T-cells prestimulated by antigen presentic cells/CD3. It is noteworthy that STAT5 is one of the transcription factors responsive to IL-7 (5157).

In this report, in addition to confirming the diabetes-suppressive properties of iDCs as we have previously published (25), we demonstrate that, at least in the NOD mouse, treatment of NOD-derived T-cells with IL-7 in vitro results in an increase in the steady-state numbers of CD4+ CD25+ T-cells in vitro. This increase is not due to proliferation but rather to the suppression of apoptosis of the CD4+ CD25+ T-cells. Furthermore, we show that IL-7 receptor α-chain (IL-7Rα) is expressed at significantly higher levels on CD4+ CD25+ T-cells compared with CD4+ CD25 cells in culture and in vivo. We also demonstrate a dramatic induction of IL-7 in diabetes-delaying iDCs. These iDCs, when injected in vivo in immunocompromised hosts reconstituted with NOD-derived T-cells, promote an increased prevalence of CD4+ CD25+ T-cells that selectively express IL-7Rα. Taken together, we propose a novel mechanism of iDC action in the suppression of type 1 diabetes onset, in which iDC-derived IL-7 prevents apoptosis of Treg populations that selectively express the α-chain of the IL-7 receptor.

Female NOD and NOD-scid mice were purchased from The Jackson Laboratories (Bar Harbor, ME) and were used between the ages of 5 and 10 weeks. They were maintained in a specific pathogen-free environment in the Animal Facility of the Rangos Research Center and used under the approval of the Animal Research Care Committee.

Generation of iDCs.

iDCs were engineered using previously published protocols (25,58). Basically, bone marrow was obtained from female NOD mice, and immature dendritic cell progenitors were propagated in vitro in granulocyte macrophage colony-stimulating factor and IL-4. At the end of the propagation, the dendritic cells were treated with a mixture of antisense oligonucleotides targeting the primary transcripts of CD40, CD80, and CD86 in which each of the antisense oligonucleotide mixtures was at a final culture concentration of 3.3 μmol/l. We use the term “iDC” to describe this engineered embodiment of NOD bone marrow–derived dendritic cells, and its immunoregulatory capability in delaying the onset of type 1 diabetes has been described previously (25).

iDC transfer to NOD mice and diabetes incidence.

Prepared as described previously (25), 2 × 106 NOD iDCs in PBS were injected by intraperitoneal route into 8-week-old female NOD mice at weekly intervals for a period of 1 month. Diabetes incidence was ascertained twice weekly in tail vein blood by electronic sampling (One-Touch; LifeScan Technologies, Milpitas, CA). Confirmation of diabetes was noted on two consecutive readings of blood glucose >280 mg/dl.

iDC-stimulated generation of Tregs in vivo.

Control dendritic cells or iDCs (1 × 105-1 × 106) from 5- to 8-week-old female NOD mice were transferred intraperitoneally into female NOD-scid mice between the ages of 5–10 weeks. Three to 5 days later, 1 × 107 column-enriched splenic T-cells (CD3+ T-cell enrichment columns; R&D Systems, Indianapolis, IN) with or without prelabeling with carboxyfluorescein ester (CFSE) (see below) were transferred through the tail vein in the same mice. One week later, splenic T-cells from the reconstituted NOD-scid mice were enriched over the T-cell columns. Cell surface phenotype of the T-cells was assessed by fluorescence-activated cell sorter (FACS) analysis in a FACSVantage SE instrument using FACSDiva and CellQuest modules (BD Biosciences, San Jose, CA).

Determination of cell proliferation and apoptosis activation.

To determine proliferation in vivo, NOD-derived enriched T-cells (female NOD mice were used at 5–8 weeks of age) were labeled in vitro with CFSE (Molecular Probes, Eugene, OR) before injection into the NOD-scid mice described immediately above. At specific times, NOD-scid mice reconstituted with the CFSE-labeled cells were killed for splenic T-cell enrichment. Proliferation was assessed by FACS analysis, measuring CFSE dilution in specific T-cell subpopulations marked by fluorescent antibodies or fluorescent reagents (like annexin V, described below). In parallel, selected T-cell populations were cocultured in vitro in the presence of control dendritic cells or iDCs followed by proliferation assessment using the CyQuant reagent (Molecular Probes) at 5 days after the initiation of coculture. Apoptosis of selected T-cell subpopulations was determined by annexin V staining/propidium iodide uptake of cells positive for defined cell surface antigens, as assessed by FACS analysis.

FACS analysis.

The following antibodies were direct conjugates of either fluorescein isothiocyanate, phycoerythrin, Cy5, CyChrome, or allophycocyanin and purchased from BD Biosciences/Pharmingen (San Diego, CA): CD4 clone, RM4-5; CD25 clone, 7D4; CD62 ligand clone, MEL-14; CD3 clone, 145-2C11; and IL-7Rα (CD127) clone, SB/199. Relevant isotype-specific, fluorescently conjugated antibodies were used throughout as controls for nonspecific cell surface binding. Cells were incubated with mixtures of specific antibodies and, in parallel, with the isotype controls at titers between 1:100 and 1:500 per the manufacturer’s suggestions. They were also labeled with propidium iodide, 7-AAD, and/or the annexin V-staining reagent and then used directly for FACS analysis. Mean fluorescence intensity was measured for cells gated on forward and side scatter properties representing T-cells. Dead cells as well as cell clumps were excluded from the analyses, which were performed using the CellQuest software package.

Effects of IL-7 on NOD-derived T-cells in vitro.

Column-enriched splenic T-cells were treated with 50 ng/ml murine recombinant IL-7 (R&D Systems) with or without 5 μg/ml concanavalin A (Sigma, St. Louis, MO) for periods between 18 and 96 h in RPMI 1640 supplemented with 10% FCS and 2 mmol/l glutamine. At the end of the specific treatment period, annexin V positivity in these T-cells was used to assess apoptosis. In parallel, the degree of proliferation in these T-cells was measured using the CyQuant fluorescence reagent (Molecular Probes). To determine whether IL-7 was directly responsible for the augmentation of CD4+ CD25+ putative Tregs in culture, we added 5 μg/ml blocking IL-7 (Peprotech, Rocky Hill, NJ) or IL-7Rα (clone A7R34) antibodies for the indicated time periods. Controls for these blocking studies were isotype-matched irrelevant antibodies (BD Biosciences/Pharmingen). In other parallel studies, column-enriched T-cells were labeled with CFSE, washed, and then treated with 50 ng/ml IL-7 with or without 5 μg/ml concanavalin A until further assay.

Detection of IL-7 in vitro.

In addition to standard RT-PCR, real-time PCR was used to ascertain the steady-state mRNA levels of IL-7 in iDC and control dendritic cell populations. Total RNA was extracted from 1 × 106–2 × 106 dendritic cells using the RNEasy kit (Qiagen, Valencia, CA). Contaminating genomic DNA was removed using RQ1 RNase-free DNase (Promega, Madison, WI). Reverse transcription was performed with 5 μg total RNA by real-time PCR using the specific primers listed further below. The mRNA levels for each of the cassettes were analyzed using the iCycler system (Bio-Rad, Hercules, CA). The mRNAs were reverse transcribed into cDNAs by using the iScript cDNA synthesis kit (Bio-Rad). Real-time PCR was performed by using a SYBR supermix kit (Bio-Rad) and running for 45 cycles (94°C for 10 s, 55°C for 10 s, and 72°C for 10 s, followed by a 5-min extension at 72°C). The mRNA level of each sample for each cassette was normalized to that of 18S RNA control PCR product (18S Housekeeping Gene; Ambion Classic II Internal Standards; Ambion, Austin, TX). Relative mRNA levels are presented as 2(Ct/β-actin − Ct/gene cassette). The primers used to amplify the mouse IL-7 cDNA product were sense, 5′-ATT ATG GGT GGT GAG AGC CG-3′, and antisense, 5′-GTT CCT GTC ATT TTG TCC AAT TCA-3′. The IL-7 and 18S RNA PCR products were also resolved in parallel in a 2% agarose gel and visualized after ethidium bromide staining.

Statistical analysis.

Student’s t test, Kaplan-Meier log-rank analysis, and ANOVA, where described, were facilitated using Prism software (version 4; GraphPad, San Diego, CA).

Administration of iDCs to NOD mice in vivo promotes the preferential survival but not the considerable proliferation of CD4+ CD25+ T-cells.

In a recent study, we showed that administration of iDCs to pre-diabetic NOD mice significantly delayed the onset of diabetes with a concomitant increase in the prevalence of CD4+ CD25+ T-cells (25). We now demonstrate that frequent administration of these dendritic cells completely prevents the onset of diabetes in NOD mice (Fig. 1A). Toward a mechanistic understanding of how the iDC administration was associated with the increased prevalence of these putative regulatory T-cells, we analyzed proliferation of CD4+ CD25+ T-cells in apoptotic and nonapoptotic states in vivo. CFSE-labeled NOD T-cells from pre-diabetic NOD female mice were injected into age- and sex-matched NOD-scid recipients that had been reconstituted with either untreated bone marrow–derived NOD dendritic cells (from 5- to 8-week-old NOD females) or iDCs 3 days before T-cell addition, as illustrated in Fig. 1B. One week later, the spleens were harvested and CD3+ T-cells were then enriched over columns. The T-cells were then stained with fluorescent antibodies to CD4 and CD25 as well as annexin V and propidium iodide. The cells were then subjected to FACS analysis as follows. First, annexin V and propidium iodide uptake levels were ascertained inside the CD4+ CD25+ gate. Each of the resulting quadrants reflecting propidium iodide/annexin V was further analyzed for CFSE dilution. The degree of CFSE peak shift and the number of peaks within each of the propidium iodide/annexin V quadrants represents the degree of proliferation of CD4+ CD25+ T-cells in the different apoptoptic states.

As a prelude to the experiments of which the data are shown in Fig. 1D and E, the general procedure followed to determine proliferation/expansion of putative Tregs or the survival of already-existing populations through ascertainment of apoptosis activation is illustrated in Fig. 1C. Starting from freshly isolated spleen, we first gated CD4+ CD25+ T-cells, and we then probed the gate for either CFSE dilution and/or annexin V positivity. Figure 1C, as a general schema, depicts the CFSE content and the annexin V positivity of CD4+ CD25+ T-cells that were cultured in the absence of IL-7 or IL-2 for a 72-h period.

To establish potential mechanisms by which iDCs lead to an augmentation of CD4+ CD25+ putative Tregs, we concurrently determined the proliferation and the incidence of apoptosis of CD4+ CD25+ T-cells obtained from NOD-scid mice previously reconstituted with iDCs and NOD splenic T-cells, the latter cells prelabeled with CFSE. Specifically, we first labeled freshly isolated NOD splenic T-cells with CFSE and subsequently injected them into NOD-scid mice that had been administered either control syngeneic dendritic cells or iDCs. Five days later the spleens of the NOD-scid recipients were excised, and enriched T-cells were subjected to FACS analysis for CFSE dilution and annexin V positivity (Fig. 1D). We gated cells around four quadrants (P2–P5), each of which represents variable CFSE dilution and annexin V positivity (and therefore variable proliferation and apoptosis; Fig. 1D). In each of these quadrants, we then ascertained the prevalence of CD4 CD25 double-positive cells (Fig. 1E). Inside the double-positive cell populations, we ultimately determined CFSE dilution (Fig. 1E).

By comparing the CFSE dilution profiles in CD4+ CD25+ cells (Fig. 1E) gated inside annexin V+/− CFSE+/− double-positive populations in the annexin V/CFSE dot plot (Fig. 1D), we propose that the degree of CFSE dilution of CD4+ CD25+ cells from control and iDC NOD-scid recipients is essentially identical. These data suggest that the augmentation of CD4+ CD25+ T-cell numbers in the iDC NOD-scid recipients was not due to expansion/proliferation of existing or progenitor cells (Fig. 1E; compare the CFSE dilution histograms in the CD4+ CD25+ cells from each of the annexin V/CFSE quadrants (P2–P5; Fig. 1D).

IL-7 increases the steady-state percentage of CD4+ CD25+ T-cells in vitro from the spleens of NOD mice.

Given the importance of IL-7 in homeostasis of T-cells, specifically with respect to survival without the need for proliferation, we speculated that our findings shown in Fig. 1E might be attributable to iDC-induced homeostatic maintenance of existing CD4+ CD25+ putative Tregs, perhaps through IL-7. We initially postulated that CD4+ CD25+ T-cells would respond identically to the effect of IL-7 as unenriched T-cell populations as previously reported (2830,33,37,40,5961). Recombinant IL-7 (50 ng/ml) exogenously added to freshly isolated NOD splenic T-cells (derived from the spleens of female, pre-diabetic NOD mice at 5–8 weeks of age) resulted in a greater prevalence of CD4+ CD25+ T-cells at 24 h after IL-7 addition. (Fig. 2A). The addition of concanavalin A led to an even greater increase in the prevalence of CD4+ CD25+ T-cells in IL-7–pretreated cells at this time over cells not stimulated with concanavalin A. The increased prevalence of CD4+ CD25+ T-cells was maintained at even 96 h after IL-7 addition with or without concanavalin A stimulation at a time when primary T-cells are expected to poorly survive in culture without the addition of IL-2 (Fig. 2B) (31,37,41,60,62).

Increased prevalence of IL-7–induced NOD CD4+ CD25+ T-cells in vitro is due to apoptosis suppression.

The observation of a considerable number of CD4+ CD25+ T-cells at 96 h after culture in the presence of IL-7 with or without concanavalin A stimulation strongly suggested a selective effect of IL-7 on survival over proliferation. We chose to examine the state of apoptosis activation by ascertaining annexin V levels in vitro. Figure 3A confirms the well-known effect of IL-7 on the survival of freshly isolated, enriched yet unfractionated T-cells (31,37,41,60,62), this time derived from the NOD strain. Figure 3B shows dramatically decreased annexin V staining of CD4+ CD25+ T-cells in IL-7–treated, freshly isolated NOD T-cell cultures with or without concanavalin A stimulation. This degree of survival was maintained at 96 h after the initiation of T-cell culture.

IL-7Rα is expressed at higher levels on NOD CD4+ CD25+ T-cells than CD4+ CD25 T-cells in vitro.

Because CD4+ CD25+ T-cells from NOD mice demonstrated significantly lower apoptosis activation as measured by annexin V staining in response to exogenous IL-7 addition, we proposed that these low-apoptotic cells would express more IL-7Rα. In Fig. 4, by FACS analysis, we show an increased level of IL-7Rα in the CD4+ CD25+ T-cell population in freshly isolated NOD splenic T-cells compared with that on CD4+ CD25 T-cells. Although concanavalin A treatment resulted in a decrease in the overall numbers of cells in both populations, the difference in the magnitude of IL-7Rα expression between CD4+ CD25+ and CD4+ CD25 T-cells remained similar.

IL-7Rα is expressed at significantly higher levels on CD4+ CD25+ T-cells compared with CD4+ CD25 T-cells in vivo.

To examine whether our in vitro observations on preferential expression of IL-7Rα on CD4+ CD25+ T-cell populations compared with levels on CD4+ CD25 held true in vivo, we reconstituted female NOD-scid mice with bone marrow–derived NOD dendritic cells and purified T-cells by sequential transfer of the dendritic cells followed 3 days after by enriched T-cells freshly isolated from Thy1.1-NOD syngeneic mice. We chose to use the Thy1.1 marker to ensure that the T-cells phenotyped in downstream analyses would, in fact, derive from donors, even in T-cell–deficient NOD-scid recipients. Furthermore, the Thy1.1 marker would also aid in normalizing the data given the predicted homeostatic expansion expected in the NOD-scid lymphopenic host. Five days after T-cell transfer, the spleens of the NOD-scid recipients were processed to obtain T-cells. In Table 1, we show increased prevalence of IL-7R+ Thy1.1+ cells in the CD4+ CD25+ population in NOD-scid hosts preconditioned with iDCs than in recipients pretreated with control dendritic cells. The prevalence of IL-7R+ Thy1.1+ cells in the CD4+ CD25 populations was very low or undetectable in iDC-preconditioned NOD-scid mice and control dendritic cell recipients, respectively (Table 1).

NOD bone marrow–derived iDCs express IL-7 in vitro.

Up to this point, our studies began suggesting to us a model whereby the levels of IL-7Rα, expressed at significantly higher levels on CD4+ CD25+ compared with CD4+ CD25 T-cells, were partly responsible for conferring to them a diabetes-delaying immunoregulatory phenotype in the NOD mouse. We further proposed that iDCs promoted the diabetes-delaying effects (25) by producing IL-7 in close proximity to, and affecting the activity of putative Tregs within, the IL-7Ra+ CD4+ CD25+ subpopulation. To address this possibility, we first looked at IL-7 production in NOD bone marrow–derived control, untreated dendritic cells and then compared it with production in iDCs. Consistent with our observations, others have previously detected IL-7 production in dendritic cells (Fig. 5A) (6366). By real-time PCR, iDCs expressed significantly higher IL-7 steady-state mRNA than did control, untreated dendritic cells (1,800 ± 265 pmol IL-7 in iDCs vs. undetectable in control dendritic cells; n = 3, data extrapolated from Ct (cycle number where fluorescence crosses the threshold) values as described in research design and methods).

To verify that it is iDC-derived IL-7 that directly promotes augmented CD4+ CD25+ putative Treg numbers and is directly involved in suppressing apoptosis activation in these cells, we repeated the coculture experiments with iDCs and freshly isolated NOD splenic T-cells in the presence of blocking IL-7 or IL-7Rα antibodies. As an additional demonstration that the cells are part of the Treg family, after the coculture, we determined the percentage of CD25+ CD62L+ cells inside a CD4+ gate (Fig. 5B) and the percentage of annexin V+ cells inside a CD4+ CD25+ CD62L+ population (Fig. 5C). The addition of blocking antibodies significantly decreased the prevalence of CD4+ CD25+ CD62L+ cells (Fig. 5B) and, moreover, blocking antibodies significantly increased apoptotic putative Tregs (Fig. 5C). Over a 96-h period, we observed a greater prevalence of dead or apoptotic cells in iDC:T-cell cocultures treated with the blocking IL-7 antibody (Fig. 5D). These data support a causal role for iDC-elaborated IL-7 in augmenting the prevalence of putative Tregs, at least in part through activation of antiapoptotic mechanisms in Tregs that express IL-7Rα. We also ascertained the cytokine production in these cocultures in the presence or absence of the blocking antibodies. With the exception of tumor necrosis factor-α and IL-6, whose levels were significantly increased in the cocultures with the blocking antibodies (Fig. 5E), the levels of all other cytokines detectable (interferon-γ, IL-2, IL-4, IL-10, transforming growth factor-β, and IL-12p70) were almost identical between control and iDC-seeded cocultures (data not shown).

IL-7Rα is constitutively expressed on the surface of NOD dendritic cells and induced at higher levels on iDCs.

We have concurrently speculated that in addition to its effects on T-cell survival, IL-7 could also promote the survival of dendritic cells in an autocrine manner. In such a putative autocrine system, dendritic cells should express IL-7Rα. As shown in Fig. 6, not only was IL-7Rα expressed on control, untreated NOD dendritic cells, but it was expressed at higher levels on iDCs.

Our previous data indicated that iDC treatment of pre-diabetic NOD mice significantly delayed diabetes onset with a single injection (25). We now demonstrate that weekly injection of iDCs for a period of 1 month commencing on the 8th week of life completely prevents diabetes in the NOD mouse. The simplistic reason could be that increased injection frequency replenished the iDCs whose lifespan is expected to be limited in vivo after exogenous administration. We had also shown that iDCs promoted an increased prevalence of CD4+ CD25+ T-cells (25). In parallel, we have observed that adoptive cotransfer of T-cells from iDC-treated, diabetes-free NOD mice along with T-cells from diabetic NOD mice into NOD-scid recipients did not result in diabetes (J.H., N.G., unpublished observations). This strengthens the argument that iDCs induce a T-cell population, possibly the same CD4+ CD25+ cells whose prevalence was increased, which somehow mediates the diabetes-suppressive effect. The maintenance of Treg numbers, in our model, would be dependent on the presence and activity of iDCs. We initially envisioned two possible mechanisms that could result in increased prevalence of the CD4+ CD25+ T-cell population: proliferation of Tregs within the CD4+ CD25+ T-cell population and/or preferential survival of this cell subpopulation in vivo. From these studies, we suggest that iDCs may not affect proliferation of CD4+ CD25+ T-cells in vivo but are responsible for maintaining their numbers, which is very likely through apoptosis suppression as evidenced by a dramatic decrease in the levels of apoptosis activation measured in annexin V/propidium iodide positivity.

Exogenous IL-7 addition to NOD T-cells resulted in preferential suppression of apoptosis activation of CD4+ CD25+ cells over CD4+ CD25 T-cells. Given the potential for regulatory activity of the former population, we propose IL-7 as an additional “costimulatory” signal promoting the preferential survival especially of putative regulatory T-cells and envisage a “coregulatory” mechanism of iDC action on these specific cell subtypes. Because our iDCs express low levels of CD40, CD80, and CD86, T-cells with TCR:antigen specific for the MHC on the iDCs would have a higher activation threshold and would be susceptible to apoptosis. An additional signal would therefore have to be supplied to suppress pro-apoptotic pathways and to permit TCR:MHC interaction time to be prolonged enough to permit adequate activation. We propose this signal to be IL-7, which would be expected to act on all T-cells with a functional IL-7 receptor coming into contact with the iDCs. Our data showing that blocking antibodies to IL-7 and IL-7Rα in iDC:T-cell cocultures increase the incidence of apoptotic CD4+ CD25+ cells and decrease their number are consistent with such a potential mechanism of action, although we do not exclude the existence of additional, complementary mechanisms that can underlie the iDC-dependent increase in Treg prevalence.

Although these data show that IL-7 is of relevance in an NOD system, we have nevertheless confirmed that IL-7 acts identically on CD4+ CD25+ putative Tregs in the C57BL/6 and Balb/c mouse (J.H., J.M., N.G., unpublished observations). Therefore, we consider it unlikely that the effects of IL-7, exogenously supplied or produced by iDCs, in promoting the augmentation of CD4+ CD25+ Tregs are NOD specific. The action of IL-7 in regulating diabetes in the NOD system via Tregs has been suggested in previous studies (67,68). Herbelin et al. (68) identified a subset of CD4+ CD62L+ T-cells able to inhibit diabetes transfer into immunodeficient NOD-scid recipients. IL-7 exhibited potentiating activity on diabetes protection via these cells that were also generated among thymocytes in vitro with exogenous IL-7 addition (68). In a subsequent study, Gombert et al. (67) reported that exogenous addition of IL-7 restored immunosuppressive IL-4 production in NOD mature thymocytes and, furthermore, that IL-7 additionally increased IL-4 production by NK1+ NOD spleen T-cells stimulated by an antibody to the CD3 complex.

Our data also suggest a method that could be useful alone or in conjunction with other published methods by which CD4+ CD25+ T-cells with regulatory activity can be expanded in vitro (22,6973). IL-7 could, therefore, be of practical utility in improving the yield, persistence in vivo, and activity of Tregs in suppressing autoimmunity or in facilitating allograft survival. Although we are excited by these novel observations suggesting IL-7 as a potential regulator/facilitator of costimulation of Tregs through suppression of apoptosis, much more work needs to be done to understand the mechanism and to translate a method into a relevant therapeutic.

FIG. 1.

Differentiation between pro-proliferative or antiapoptotic effects of iDCs on T-cell subsets. A: Frequent iDC administration to pre-diabetic NOD mice completely prevents diabetes. Weekly injections of 2 × 106 iDCs into NOD female recipients beginning at 8 weeks of age for a period of 1 month resulted in all recipients being diabetes free at >32 weeks of age. P < 0.0001 by Kaplan-Meier log-rank analysis. B: Reconstitution of NOD-scid mice with immune cell populations. To determine the effects of the iDCs in vivo, we first injected 1 × 106–2 × 106 dendritic cell embodiments (control dendritic cells or iDCs) into 5- to 8-week-old NOD-scid female mice. In parallel, column-enriched splenic T-cells from age-matched female NOD mice were labeled in culture with CFSE. Three days later, 1 × 107 of the CFSE-labeled column-enriched NOD splenic T-cells were injected into the NOD-scid recipients by tail vein injection. These “incubator mice” were killed 1 week later, and the spleens were collected for T-cell enrichment, processing, and downstream FACS analysis. C: General schema of FACS analysis. This panel describes the strategy for differentiating between proliferation and apoptosis activation in selected T-cell subsets. After staining with fluorescent label–coupled antibodies to CD4 and CD25, cells representing T-cells were first gated (top left panel; P1) on forward and side scatter. CD4+ CD25+ double-positive cells were then isolated inside a new gate (middle left panel; P2). Within this gate, the dilution of CFSE (top right panel) or the level of annexin V (bottom right panel) was ascertained. D: Freshly isolated splenic T-cells from NOD-scid mice reconstituted with dendritic cells and NOD T-cells; gating into proliferative and apoptotic double-positive populations. Freshly isolated splenic T-cells from NOD-scid mice reconstituted with dendritic cells and NOD T-cells as described (top panel; cells selected for subsequent FACS analyses) were assigned into one of four FACS gates based on CFSE dilution and annexin V positivity: P2, proliferating and apoptotic; P3, nonproliferating and apoptotic; P4, proliferating and low apoptotic; and P5, nonproliferating and low apoptotic. Each of these gates was then probed for the analysis presented in E. E: iDCs do not promote discernible proliferation of CD4+ CD25+ T-cells compared with control dendritic cells in vivo. Comparing the CFSE dilution among CD4+ CD25+ T-cells from iDCs and control dendritic cell–preconditioned NOD-scid incubator mice, it is evident that negligible proliferation occurred in vivo. The CFSE dilution reflects the proliferation of the CD4+ CD25+ T-cells inside the NOD-scid incubator mice. CFSE-prelabeled splenic T-cells from incubator NOD-scid mice were processed for annexin V and then stained with fluorescence-coupled antibodies to CD4 and CD25. D shows how the cells were first selected based on annexin V and CFSE characteristics. The quadrants in E illustrate the CD4 CD25 cells within each of the quadrants shown in D (P2–P5). The CFSE dilution histograms in this panel refer to the CD4+ CD25+ double-positive quadrants shown herein. These data are representative of the prevalence of each cell population as observed in freshly isolated T-cells from three different NOD-scid mice (iDC-preconstituted mice n = 3; control dendritic cell–preconstituted mice n = 3).

FIG. 1.

Differentiation between pro-proliferative or antiapoptotic effects of iDCs on T-cell subsets. A: Frequent iDC administration to pre-diabetic NOD mice completely prevents diabetes. Weekly injections of 2 × 106 iDCs into NOD female recipients beginning at 8 weeks of age for a period of 1 month resulted in all recipients being diabetes free at >32 weeks of age. P < 0.0001 by Kaplan-Meier log-rank analysis. B: Reconstitution of NOD-scid mice with immune cell populations. To determine the effects of the iDCs in vivo, we first injected 1 × 106–2 × 106 dendritic cell embodiments (control dendritic cells or iDCs) into 5- to 8-week-old NOD-scid female mice. In parallel, column-enriched splenic T-cells from age-matched female NOD mice were labeled in culture with CFSE. Three days later, 1 × 107 of the CFSE-labeled column-enriched NOD splenic T-cells were injected into the NOD-scid recipients by tail vein injection. These “incubator mice” were killed 1 week later, and the spleens were collected for T-cell enrichment, processing, and downstream FACS analysis. C: General schema of FACS analysis. This panel describes the strategy for differentiating between proliferation and apoptosis activation in selected T-cell subsets. After staining with fluorescent label–coupled antibodies to CD4 and CD25, cells representing T-cells were first gated (top left panel; P1) on forward and side scatter. CD4+ CD25+ double-positive cells were then isolated inside a new gate (middle left panel; P2). Within this gate, the dilution of CFSE (top right panel) or the level of annexin V (bottom right panel) was ascertained. D: Freshly isolated splenic T-cells from NOD-scid mice reconstituted with dendritic cells and NOD T-cells; gating into proliferative and apoptotic double-positive populations. Freshly isolated splenic T-cells from NOD-scid mice reconstituted with dendritic cells and NOD T-cells as described (top panel; cells selected for subsequent FACS analyses) were assigned into one of four FACS gates based on CFSE dilution and annexin V positivity: P2, proliferating and apoptotic; P3, nonproliferating and apoptotic; P4, proliferating and low apoptotic; and P5, nonproliferating and low apoptotic. Each of these gates was then probed for the analysis presented in E. E: iDCs do not promote discernible proliferation of CD4+ CD25+ T-cells compared with control dendritic cells in vivo. Comparing the CFSE dilution among CD4+ CD25+ T-cells from iDCs and control dendritic cell–preconditioned NOD-scid incubator mice, it is evident that negligible proliferation occurred in vivo. The CFSE dilution reflects the proliferation of the CD4+ CD25+ T-cells inside the NOD-scid incubator mice. CFSE-prelabeled splenic T-cells from incubator NOD-scid mice were processed for annexin V and then stained with fluorescence-coupled antibodies to CD4 and CD25. D shows how the cells were first selected based on annexin V and CFSE characteristics. The quadrants in E illustrate the CD4 CD25 cells within each of the quadrants shown in D (P2–P5). The CFSE dilution histograms in this panel refer to the CD4+ CD25+ double-positive quadrants shown herein. These data are representative of the prevalence of each cell population as observed in freshly isolated T-cells from three different NOD-scid mice (iDC-preconstituted mice n = 3; control dendritic cell–preconstituted mice n = 3).

Close modal
FIG. 2.

A: IL-7 promotes survival of CD4+ CD25+ NOD splenic T-cells in vitro. Increased number of CD4+ CD25+ T-cells is observed after a 24-h exposure to 50 ng/ml IL-7 in culture. Freshly isolated splenocytes from 5- to 8-week-old NOD mice were column enriched and cultured in the presence or absence of IL-7, with or without concanavalin A stimulation (5 μg/ml). Twenty-four hours later, the cells were stained with fluorescence-coupled antibodies to CD4 and CD25 and processed for FACS analysis. At the top of the panel, P1 shows the cells selected for FACS analysis based on forward and side scatter properties associated with T-cells. Each of the FACS plots represents CD4 and CD25 cells. These data are representative of four independently performed experiments. B: IL-7 maintains CD4+ CD25+ T-cell numbers at 96 h after exposure in vitro. CD4+ CD25+ cell numbers observed at 24 h after IL-7 exposure were maintained at 96 h in vitro. Freshly isolated splenocytes from 5- to 8-week-old NOD mice were column enriched and cultured in the presence or absence of IL-7, with or without concanavalin A stimulation (5 μg/ml) for 5 days. FACS analysis for cell prevalence was performed identically as above. These data are representative of four independently performed experiments.

FIG. 2.

A: IL-7 promotes survival of CD4+ CD25+ NOD splenic T-cells in vitro. Increased number of CD4+ CD25+ T-cells is observed after a 24-h exposure to 50 ng/ml IL-7 in culture. Freshly isolated splenocytes from 5- to 8-week-old NOD mice were column enriched and cultured in the presence or absence of IL-7, with or without concanavalin A stimulation (5 μg/ml). Twenty-four hours later, the cells were stained with fluorescence-coupled antibodies to CD4 and CD25 and processed for FACS analysis. At the top of the panel, P1 shows the cells selected for FACS analysis based on forward and side scatter properties associated with T-cells. Each of the FACS plots represents CD4 and CD25 cells. These data are representative of four independently performed experiments. B: IL-7 maintains CD4+ CD25+ T-cell numbers at 96 h after exposure in vitro. CD4+ CD25+ cell numbers observed at 24 h after IL-7 exposure were maintained at 96 h in vitro. Freshly isolated splenocytes from 5- to 8-week-old NOD mice were column enriched and cultured in the presence or absence of IL-7, with or without concanavalin A stimulation (5 μg/ml) for 5 days. FACS analysis for cell prevalence was performed identically as above. These data are representative of four independently performed experiments.

Close modal
FIG. 3.

A: Exogenous IL-7 addition to NOD T-cells reduces apoptosis activation in vitro. Significant decrease in the number of annexin V+ CD4+ CD25+ T-cells after IL-7 treatment of NOD T-cells reflects an effect of IL-7 on apoptosis suppression. Freshly isolated splenocytes from 5- to 8-week-old NOD mice were column enriched and cultured in the presence or absence of IL-7, with or without concanavalin A stimulation (5 μg/ml). Twenty-four hours later, the cells were stained with fluorescence-coupled antibodies to CD4 and CD25, stained for annexin V, and processed for FACS analysis. CD4+ CD25+ cells were gated and then examined for annexin V positivity. Each of the histograms indicates the level of apoptosis activation. These data are representative of four independently performed experiments. B: IL-7 maintains suppressed apoptosis activation of CD4+ CD25+ T-cell numbers at 96 h after exposure in vitro. The levels of annexin V in CD4+ CD25+ cells observed at 24 h after IL-7 exposure were maintained or significantly improved at 96 h in vitro. Freshly isolated splenocytes from 5- to 8-week-old NOD mice were column enriched and cultured in the presence or absence of IL-7, with or without concanavalin A stimulation (5 μg/ml) for 5 days. FACS analysis for annexin V prevalence in the CD4+ CD25+ cell population was performed identically as above. These data are representative of four independently performed experiments.

FIG. 3.

A: Exogenous IL-7 addition to NOD T-cells reduces apoptosis activation in vitro. Significant decrease in the number of annexin V+ CD4+ CD25+ T-cells after IL-7 treatment of NOD T-cells reflects an effect of IL-7 on apoptosis suppression. Freshly isolated splenocytes from 5- to 8-week-old NOD mice were column enriched and cultured in the presence or absence of IL-7, with or without concanavalin A stimulation (5 μg/ml). Twenty-four hours later, the cells were stained with fluorescence-coupled antibodies to CD4 and CD25, stained for annexin V, and processed for FACS analysis. CD4+ CD25+ cells were gated and then examined for annexin V positivity. Each of the histograms indicates the level of apoptosis activation. These data are representative of four independently performed experiments. B: IL-7 maintains suppressed apoptosis activation of CD4+ CD25+ T-cell numbers at 96 h after exposure in vitro. The levels of annexin V in CD4+ CD25+ cells observed at 24 h after IL-7 exposure were maintained or significantly improved at 96 h in vitro. Freshly isolated splenocytes from 5- to 8-week-old NOD mice were column enriched and cultured in the presence or absence of IL-7, with or without concanavalin A stimulation (5 μg/ml) for 5 days. FACS analysis for annexin V prevalence in the CD4+ CD25+ cell population was performed identically as above. These data are representative of four independently performed experiments.

Close modal
FIG. 4.

CD4+ CD25+ T-cells from NOD mice express higher cell surface IL-7 receptor α-chain than CD4+ CD25 cells. Freshly isolated T-cells from 5- to 8-week-old NOD female mice exhibit higher IL-7 receptor α-chain in the CD4+ CD25+ population than in the CD4+ CD25 population. Column-enriched T-cells were stained with fluorescence-coupled antibodies to CD4, to CD25, and to the α-chain of the IL-7 receptor. FACS analysis was performed on cells with a forward/side scatter profile indicative of T-cells, and the levels of IL-7Rα were ascertained inside each of the quadrants in the CD4 CD25 subgate. These data are representative of three independent T-cell isolations.

FIG. 4.

CD4+ CD25+ T-cells from NOD mice express higher cell surface IL-7 receptor α-chain than CD4+ CD25 cells. Freshly isolated T-cells from 5- to 8-week-old NOD female mice exhibit higher IL-7 receptor α-chain in the CD4+ CD25+ population than in the CD4+ CD25 population. Column-enriched T-cells were stained with fluorescence-coupled antibodies to CD4, to CD25, and to the α-chain of the IL-7 receptor. FACS analysis was performed on cells with a forward/side scatter profile indicative of T-cells, and the levels of IL-7Rα were ascertained inside each of the quadrants in the CD4 CD25 subgate. These data are representative of three independent T-cell isolations.

Close modal
FIG. 5.

A: iDCs, but not control dendritic cells from NOD mice, express IL-7. RT-PCR demonstrates that iDCs express detectable steady-state mRNA for IL-7 but that control, untreated dendritic cells do not. PCR products from cDNA were loaded and separated in a 2% agarose gel, stained with ethidium bromide, and visualized under ultraviolet light. PCR was performed for 45 cycles. As a positive control and to equalize the amount of RNA analyzed, 18S RNA was used as an internal standard. Dendritic cells propagated from bone marrow of five different pre-diabetic NOD mice and treated with the oligonucleotides all demonstrated IL-7 steady-state mRNA as shown, but none of five untreated dendritic cells from the same mice expressed IL-7 message. B: Blocking antibodies to IL-7 or IL-7Rα decrease the prevalence of CD4+ CD25+ T-cells in iDC:T-cell cocultures. Shown are the FACS data of CD25+ CD62L+ cells inside a CD4 gate, where the cells derive from dendritic cell:T-cell cocultures at the end of 5 days. AS-DC, antisense oligonucleotide–treated NOD dendritic cell; IL-7(BL), blocking antibody to the IL-7 cytokine; IL-7(isotype), IL-7 isotype–matched irrelevant antibody; IL-7Rα(BL), blocking antibody to the IL-7R α-chain; IL-7Rα(isotype), IL-7Rα isotype–matched irrelevant antibody; Tc, T-cells from freshly isolated NOD spleen. These data are representative of three independent experiments, each performed in triplicate. The error bars represent the SE. P < 0.01 between control cocultures and those in which blocking antibodies were added (ANOVA). C: Blocking antibodies to IL-7 or IL-7Rα increase the prevalence of apoptotic CD4+ CD25+ T-cells in iDC:T-cell cocultures. Shown are the FACSdata of annexin V+ cells inside a CD4+ CD25+ CD62L+ gate, where the cells derive from dendritic cell:T-cell cocultures at the end of 5 days. These data are representative of three independent experiments, each performed in triplicate. The error bars represent the SE. P < 0.05 between control cocultures and those in which blocking antibodies were added (ANOVA). D: Prevalence of apoptotic and dead CD4+ CD25+ CD62L+ T-cells derived from iDC:T-cell cocultures increases over time in the presence of blocking IL-7 antibodies. T-cells from the cocultures were first gated into CD4+ cells, and within this gate, CD25+ CD62L+ cells were then gated. Propidium iodide+ annexin V+ double-positive cells were determined inside this putative Treg population. Cells were removed from cocultures at the end of 24, 72, and 96 h incubation. These data are representative of two independent experiments in triplicate, and the error bars represent the SE. P < 0.001 between control cocultures and those treated with the blocking IL-7 antibody, at all time points (ANOVA). E: Blocking antibodies to IL-7 or IL-7Rα increases the production of tumor necrosis factor-α and IL-6 in iDC:T-cell cocultures. Cytokine production was assessed in coculture supernatant at the end of a 5-day incubation. These data are representative of three independent experiments, each performed in triplicate. The error bars represent the SE. P < 0.05 between control cocultures and those in which blocking antibodies were added (ANOVA).

FIG. 5.

A: iDCs, but not control dendritic cells from NOD mice, express IL-7. RT-PCR demonstrates that iDCs express detectable steady-state mRNA for IL-7 but that control, untreated dendritic cells do not. PCR products from cDNA were loaded and separated in a 2% agarose gel, stained with ethidium bromide, and visualized under ultraviolet light. PCR was performed for 45 cycles. As a positive control and to equalize the amount of RNA analyzed, 18S RNA was used as an internal standard. Dendritic cells propagated from bone marrow of five different pre-diabetic NOD mice and treated with the oligonucleotides all demonstrated IL-7 steady-state mRNA as shown, but none of five untreated dendritic cells from the same mice expressed IL-7 message. B: Blocking antibodies to IL-7 or IL-7Rα decrease the prevalence of CD4+ CD25+ T-cells in iDC:T-cell cocultures. Shown are the FACS data of CD25+ CD62L+ cells inside a CD4 gate, where the cells derive from dendritic cell:T-cell cocultures at the end of 5 days. AS-DC, antisense oligonucleotide–treated NOD dendritic cell; IL-7(BL), blocking antibody to the IL-7 cytokine; IL-7(isotype), IL-7 isotype–matched irrelevant antibody; IL-7Rα(BL), blocking antibody to the IL-7R α-chain; IL-7Rα(isotype), IL-7Rα isotype–matched irrelevant antibody; Tc, T-cells from freshly isolated NOD spleen. These data are representative of three independent experiments, each performed in triplicate. The error bars represent the SE. P < 0.01 between control cocultures and those in which blocking antibodies were added (ANOVA). C: Blocking antibodies to IL-7 or IL-7Rα increase the prevalence of apoptotic CD4+ CD25+ T-cells in iDC:T-cell cocultures. Shown are the FACSdata of annexin V+ cells inside a CD4+ CD25+ CD62L+ gate, where the cells derive from dendritic cell:T-cell cocultures at the end of 5 days. These data are representative of three independent experiments, each performed in triplicate. The error bars represent the SE. P < 0.05 between control cocultures and those in which blocking antibodies were added (ANOVA). D: Prevalence of apoptotic and dead CD4+ CD25+ CD62L+ T-cells derived from iDC:T-cell cocultures increases over time in the presence of blocking IL-7 antibodies. T-cells from the cocultures were first gated into CD4+ cells, and within this gate, CD25+ CD62L+ cells were then gated. Propidium iodide+ annexin V+ double-positive cells were determined inside this putative Treg population. Cells were removed from cocultures at the end of 24, 72, and 96 h incubation. These data are representative of two independent experiments in triplicate, and the error bars represent the SE. P < 0.001 between control cocultures and those treated with the blocking IL-7 antibody, at all time points (ANOVA). E: Blocking antibodies to IL-7 or IL-7Rα increases the production of tumor necrosis factor-α and IL-6 in iDC:T-cell cocultures. Cytokine production was assessed in coculture supernatant at the end of a 5-day incubation. These data are representative of three independent experiments, each performed in triplicate. The error bars represent the SE. P < 0.05 between control cocultures and those in which blocking antibodies were added (ANOVA).

Close modal
FIG. 6.

Detectable cell surface IL-7Rα on NOD dendritic cells and increased level on iDCs. FACS analysis demonstrates detectable levels of IL-7R α-chain on the surface of bone marrow–derived dendritic cells from NOD mice (5- to 8-week-old females) and increased in iDCs. The cells were produced in vitro as described in research design and methods. They were then stained with a fluorescence-coupled antibody to the α-chain of IL-7R and processed for FACS. These data are representative of measurements made in three independently propagated dendritic cell cultures.

FIG. 6.

Detectable cell surface IL-7Rα on NOD dendritic cells and increased level on iDCs. FACS analysis demonstrates detectable levels of IL-7R α-chain on the surface of bone marrow–derived dendritic cells from NOD mice (5- to 8-week-old females) and increased in iDCs. The cells were produced in vitro as described in research design and methods. They were then stained with a fluorescence-coupled antibody to the α-chain of IL-7R and processed for FACS. These data are representative of measurements made in three independently propagated dendritic cell cultures.

Close modal
TABLE 1

NOD-scid mice preconstituted with dendritic cell embodiments

IL-7Rα+ Thyl.1+
Control dendritic cells (n = 4) (%)  
    Gated CD4+ CD25+ cells 2.2 ± 1.9 
    Gated CD4+ CD25 cells ND 
iDCs (n = 4) (%)  
    Gated CD4+ CD25+ cells 33.3 ± 8.6 
    Gated CD4+ CD25 cells 8.9 ± 5.0 
IL-7Rα+ Thyl.1+
Control dendritic cells (n = 4) (%)  
    Gated CD4+ CD25+ cells 2.2 ± 1.9 
    Gated CD4+ CD25 cells ND 
iDCs (n = 4) (%)  
    Gated CD4+ CD25+ cells 33.3 ± 8.6 
    Gated CD4+ CD25 cells 8.9 ± 5.0 

Data are means ± SE. iDCs promote an increase in IL-7Rα+ CD4+ CD25+ T-cells in vivo. Compared with CD4+ CD25 cells, IL-7Rα is found at considerably higher levels at the surface of CD4+ CD25+ cells and, more importantly, in iDC-preconditioned NOD-scid recipients compared with those administered control dendritic cells. Five- to 8-week-old NOD-scid female mice were first injected with 1 × 106 iDCs or control dendritic cells (n = 4). Three days later, column-enriched splenocyte-derived T-cells from Thy1.1+ NOD age-matched female mice were injected into the NOD-scid recipients. Five days after the T-cell transfer, the spleens of the NOD-scid mice were isolated, and single cells were column enriched. The T-cells were stained with fluorescence-coupled antibodies to Thy1.1, IL-7Rα, CD4, and CD25. Relevant fluorescence-conjugated isotype controls were used as controls throughout. The cells were gated identically as shown in Fig. 4, with the additional subgating of cells into quadrants of IL-7Rα and Thy1.1. The table shows representative data from four mice per reconstitution group; each experiment was performed twice. P < 0.001, prevalence of IL-7Rα+ Thy1.1+ cells in the gated CD4+CD25+ population between control and iDC groups (ANOVA). P < 0.001, prevalence of IL-7Rα+ Thy1.1+cells between CD4+ CD25+ and CD4+ CD25 cells irrespective of control dendritic cell or iDC preconstitution (ANOVA). ND, not detected.

N.G. has received National Institutes of Health Grant DK-49835. M.T. has received National Institutes of Health Grant DK-063499 and Juvenile Diabetes Research Foundation Program Project 4-1999-845.

We thank Dr. Henry Dong for critical reading of the manuscript.

1.
Thomson AW: Designer dendritic cells for transplant tolerance.
Transplant Proc
34
:
2727
–2728,
2002
2.
Coates PT, Thomson AW: Dendritic cells, tolerance induction and transplant outcome.
Am J Transplant
2
:
299
–307,
2002
3.
Morelli AE, Hackstein H, Thomson AW: Potential of tolerogenic dendritic cells for transplantation.
Semin Immunol
13
:
323
–335,
2001
4.
Hackstein H, Morelli AE, Thomson AW: Designer dendritic cells for tolerance induction: guided not misguided missiles.
Trends Immunol
22
:
437
–442,
2001
5.
Banchereau J, Steinman RM: Dendritic cells and the control of immunity.
Nature
392
:
245
–252,
1998
6.
Lu L, Qian S, Hershberger PA, Rudert WA, Lynch DH, Thomson AW: Fas ligand (CD95L) and B7 expression on dendritic cells provide counter-regulatory signals for T cell survival and proliferation.
J Immunol
158
:
5676
–5684,
1997
7.
Coates PT, Colvin BL, Kaneko K, Taner T, Thomson AW: Pharmacologic, biologic, and genetic engineering approaches to potentiation of donor-derived dendritic cell tolerogenicity.
Transplantation
75
:
32S
–36S,
2003
8.
Mellor AL, Munn DH: IDO expression by dendritic cells: tolerance and tryptophan catabolism.
Nat Rev Immunol
4
:
762
–774,
2004
9.
Mellor AL, Chandler P, Baban B, Hansen AM, Marshall B, Pihkala J, Waldmann H, Cobbold S, Adams E, Munn DH: Specific subsets of murine dendritic cells acquire potent T cell regulatory functions following CTLA4-mediated induction of indoleamine 2,3 dioxygenase.
Int Immunol
16
:
1391
–1401,
2004
10.
Munn DH, Sharma MD, Hou D, Baban B, Lee JR, Antonia SJ, Messina JL, Chandler P, Koni PA, Mellor AL: Expression of indoleamine 2,3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes.
J Clin Invest
114
:
280
–290,
2004
11.
Munn DH, Sharma MD, Mellor AL: Ligation of B7-1/B7-2 by human CD4+ T cells triggers indoleamine 2,3-dioxygenase activity in dendritic cells.
J Immunol
172
:
4100
–4110,
2004
12.
Mellor AL, Baban B, Chandler P, Marshall B, Jhaver K, Hansen A, Koni PA, Iwashima M, Munn DH: Cutting edge: induced indoleamine 2,3 dioxygenase expression in dendritic cell subsets suppresses T cell clonal expansion.
J Immunol
171
:
1652
–1655,
2003
13.
Munn DH, Sharma MD, Lee JR, Jhaver KG, Johnson TS, Keskin DB, Marshall B, Chandler P, Antonia SJ, Burgess R, Slingluff CL Jr, Mellor AL: Potential regulatory function of human dendritic cells expressing indoleamine 2,3-dioxygenase.
Science
297
:
1867
–1870,
2002
14.
Fallarino F, Vacca C, Orabona C, Belladonna ML, Bianchi R, Marshall B, Keskin DB, Mellor AL, Fioretti MC, Grohmann U, Puccetti P: Functional expression of indoleamine 2,3-dioxygenase by murine CD8 alpha(+) dendritic cells.
Int Immunol
14
:
65
–68,
2002
15.
Fan Z, Huang XL, Borowski L, Mellors JW, Rinaldo CR Jr: Restoration of anti-human immunodeficiency virus type 1 (HIV-1) responses in CD8+ T cells from late-stage patients on prolonged antiretroviral therapy by stimulation in vitro with HIV-1 protein-loaded dendritic cells.
J Virol
75
:
4413
–4419,
2001
16.
Yamazaki S, Iyoda T, Tarbell K, Olson K, Velinzon K, Inaba K, Steinman RM: Direct expansion of functional CD25+ CD4+ regulatory T cells by antigen-processing dendritic cells.
J Exp Med
198
:
235
–247,
2003
17.
Tarbell KV, Yamazaki S, Olson K, Toy P, Steinman RM: CD25+ CD4+ T cells, expanded with dendritic cells presenting a single autoantigenic peptide, suppress autoimmune diabetes.
J Exp Med
199
:
1467
–1477,
2004
18.
Kubo T, Hatton RD, Oliver J, Liu X, Elson CO, Weaver CT: Regulatory T cell suppression and anergy are differentially regulated by proinflammatory cytokines produced by TLR-activated dendritic cells.
J Immunol
173
:
7249
–7258,
2004
19.
Gad M, Kristensen NN, Kury E, Claesson MH: Characterization of T-regulatory cells, induced by immature dendritic cells, which inhibit enteroantigen-reactive colitis-inducing T-cell responses in vitro and in vivo.
Immunology
113
:
499
–508,
2004
20.
Moseman EA, Liang X, Dawson AJ, Panoskaltsis-Mortari A, Krieg AM, Liu YJ, Blazar BR, Chen W: Human plasmacytoid dendritic cells activated by CpG oligodeoxynucleotides induce the generation of CD4+CD25+ regulatory T cells.
J Immunol
173
:
4433
–4442,
2004
21.
Rutella S, Lemoli RM: Regulatory T cells and tolerogenic dendritic cells: from basic biology to clinical applications.
Immunol Lett
94
:
11
–26,
2004
22.
Godfrey WR, Ge YG, Spoden DJ, Levine BL, June CH, Blazar BR, Porter SB: In vitro-expanded human CD4(+)CD25(+) T-regulatory cells can markedly inhibit allogeneic dendritic cell-stimulated MLR cultures.
Blood
104
:
453
–461,
2004
23.
Oldenhove G, de Heusch M, Urbain-Vansanten G, Urbain J, Maliszewski C, Leo O, Moser M: CD4+ CD25+ regulatory T cells control T helper cell type 1 responses to foreign antigens induced by mature dendritic cells in vivo.
J Exp Med
198
:
259
–266,
2003
24.
Min WP, Zhou D, Ichim TE, Strejan GH, Xia X, Yang J, Huang X, Garcia B, White D, Dutartre P, Jevnikar AM, Zhong R: Inhibitory feedback loop between tolerogenic dendritic cells and regulatory T cells in transplant tolerance.
J Immunol
170
:
1304
–1312,
2003
25.
Machen J, Harnaha J, Lakomy R, Styche A, Trucco M, Giannoukakis N: Antisense oligonucleotides down-regulating costimulation confer diabetes-preventive properties to nonobese diabetic mouse dendritic cells.
J Immunol
173
:
4331
–4341,
2004
26.
Fry TJ, Mackall CL: Interleukin-7: from bench to clinic.
Blood
99
:
3892
–3904,
2002
27.
Fry TJ, Connick E, Falloon J, Lederman MM, Liewehr DJ, Spritzler J, Steinberg SM, Wood LV, Yarchoan R, Zuckerman J, Landay A, Mackall CL: A potential role for interleukin-7 in T-cell homeostasis.
Blood
97
:
2983
–2990,
2001
28.
Fry TJ, Mackall CL: Interleukin-7: master regulator of peripheral T-cell homeostasis?
Trends Immunol
22
:
564
–571,
2001
29.
Jaleco S, Kinet S, Hassan J, Dardalhon V, Swainson L, Reen D, Taylor N: IL-7 and CD4+ T-cell proliferation.
Blood
100
:
4676
–4677 [author reply 4677–4678,
2002
]
30.
Geiselhart LA, Humphries CA, Gregorio TA, Mou S, Subleski J, Komschlies KL: IL-7 administration alters the CD4:CD8 ratio, increases T cell numbers, and increases T cell function in the absence of activation.
J Immunol
166
:
3019
–3027,
2001
31.
Gringhuis SI, de Leij LF, Verschuren EW, Borger P, Vellenga E: Interleukin-7 upregulates the interleukin-2-gene expression in activated human T lymphocytes at the transcriptional level by enhancing the DNA binding activities of both nuclear factor of activated T cells and activator protein-1.
Blood
90
:
2690
–2700,
1997
32.
Costello R, Brailly H, Mallet F, Mawas C, Olive D: Interleukin-7 is a potent co-stimulus of the adhesion pathway involving CD2 and CD28 molecules.
Immunology
80
:
451
–457,
1993
33.
Chazen GD, Pereira GM, LeGros G, Gillis S, Shevach EM: Interleukin 7 is a T-cell growth factor.
Proc Natl Acad Sci U S A
86
:
5923
–5927,
1989
34.
Borger P, Kauffman HF, Postma DS, Vellenga E: IL-7 differentially modulates the expression of IFN-gamma and IL-4 in activated human T lymphocytes by transcriptional and post-transcriptional mechanisms.
J Immunol
156
:
1333
–1338,
1996
35.
Mehrotra PT, Grant AJ, Siegel JP: Synergistic effects of IL-7 and IL-12 on human T cell activation.
J Immunol
154
:
5093
–5102,
1995
36.
Schober SL, Kuo CT, Schluns KS, Lefrancois L, Leiden JM, Jameson SC: Expression of the transcription factor lung Kruppel-like factor is regulated by cytokines and correlates with survival of memory T cells in vitro and in vivo.
J Immunol
163
:
3662
–3667,
1999
37.
Lynch DH, Miller RE: Interleukin 7 promotes long-term in vitro growth of antitumor cytotoxic T lymphocytes with immunotherapeutic efficacy in vivo.
J Exp Med
179
:
31
–42,
1994
38.
Ku CC, Murakami M, Sakamoto A, Kappler J, Marrack P: Control of homeostasis of CD8+ memory T cells by opposing cytokines.
Science
288
:
675
–678,
2000
39.
Fry TJ, Christensen BL, Komschlies KL, Gress RE, Mackall CL: Interleukin-7 restores immunity in athymic T-cell-depleted hosts.
Blood
97
:
1525
–1533,
2001
40.
Boise LH, Minn AJ, June CH, Lindsten T, Thompson CB: Growth factors can enhance lymphocyte survival without committing the cell to undergo cell division.
Proc Natl Acad Sci U S A
92
:
5491
–5495,
1995
41.
Vella AT, Dow S, Potter TA, Kappler J, Marrack P: Cytokine-induced survival of activated T cells in vitro and in vivo.
Proc Natl Acad Sci U S A
95
:
3810
–3815,
1998
42.
Furtado GC, Curotto de Lafaille MA, Kutchukhidze N, Lafaille JJ: Interleukin 2 signaling is required for CD4(+) regulatory T cell function.
J Exp Med
196
:
851
–857,
2002
43.
Almeida AR, Legrand N, Papiernik M, Freitas AA: Homeostasis of peripheral CD4+ T cells: IL-2R alpha and IL-2 shape a population of regulatory cells that controls CD4+ T cell numbers.
J Immunol
169
:
4850
–4860,
2002
44.
Van Parijs L, Refaeli Y, Lord JD, Nelson BH, Abbas AK, Baltimore D: Uncoupling IL-2 signals that regulate T cell proliferation, survival, and Fas-mediated activation-induced cell death.
Immunity
11
:
281
–288,
1999
45.
Nelson BH, Willerford DM: Biology of the interleukin-2 receptor.
Adv Immunol
70
:
1
–81,
1998
46.
Moriggl R, Topham DJ, Teglund S, Sexl V, McKay C, Wang D, Hoffmeyer A, van Deursen J, Sangster MY, Bunting KD, Grosveld GC, Ihle JN: Stat5 is required for IL-2-induced cell cycle progression of peripheral T cells.
Immunity
10
:
249
–259,
1999
47.
Moriggl R, Sexl V, Piekorz R, Topham D, Ihle JN: Stat5 activation is uniquely associated with cytokine signaling in peripheral T cells.
Immunity
11
:
225
–230,
1999
48.
Snow JW, Abraham N, Ma MC, Herndier BG, Pastuszak AW, Goldsmith MA: Loss of tolerance and autoimmunity affecting multiple organs in STAT5A/5B-deficient mice.
J Immunol
171
:
5042
–5050,
2003
49.
Burchill MA, Goetz CA, Prlic M, O’Neil JJ, Harmon IR, Bensinger SJ, Turka LA, Brennan P, Jameson SC, Farrar MA: Distinct effects of STAT5 activation on CD4+ and CD8+ T cell homeostasis: development of CD4+CD25+ regulatory T cells versus CD8+ memory T cells.
J Immunol
171
:
5853
–5864,
2003
50.
Goetz CA, Harmon IR, O’Neil JJ, Burchill MA, Farrar MA: STAT5 activation underlies IL7 receptor-dependent B cell development.
J Immunol
172
:
4770
–4778,
2004
51.
Stanton ML, Brodeur PH: Stat5 mediates the IL-7-induced accessibility of a representative D-Distal VH gene.
J Immunol
174
:
3164
–3168,
2005
52.
Xue HH, Kovanen PE, Pise-Masison CA, Berg M, Radovich MF, Brady JN, Leonard WJ: IL-2 negatively regulates IL-7 receptor alpha chain expression in activated T lymphocytes.
Proc Natl Acad Sci U S A
99
:
13759
–13764,
2002
53.
Ye SK, Agata Y, Lee HC, Kurooka H, Kitamura T, Shimizu A, Honjo T, Ikuta K: The IL-7 receptor controls the accessibility of the TCRgamma locus by Stat5 and histone acetylation.
Immunity
15
:
813
–823,
2001
54.
Sato AK, Yanai N, Okubo T, Mori KJ, Obinata M: Stromal cells provide signals different from cytokines for STAT5 activation in hematopoietic cells.
Cell Struct Funct
26
:
95
–101,
2001
55.
Leonard WJ, Imada K, Nakajima H, Puel A, Soldaini E, John S: Signaling via the IL-2 and IL-7 receptors from the membrane to the nucleus.
Cold Spring Harb Symp Quant Biol
64
:
417
–424,
1999
56.
Rosenthal LA, Winestock KD, Finbloom DS: IL-2 and IL-7 induce heterodimerization of STAT5 isoforms in human peripheral blood T lymphoblasts.
Cell Immunol
181
:
172
–181,
1997
57.
Foxwell BM, Beadling C, Guschin D, Kerr I, Cantrell D: Interleukin-7 can induce the activation of Jak 1, Jak 3 and STAT 5 proteins in murine T cells.
Eur J Immunol
25
:
3041
–3046,
1995
58.
Giannoukakis N, Bonham CA, Qian S, Zhou Z, Peng L, Harnaha J, Li W, Thomson AW, Fung JJ, Robbins PD, Lu L: Prolongation of cardiac allograft survival using dendritic cells treated with NF-kappaB decoy oligodeoxyribonucleotides.
Mol Ther
1
:
430
–437,
2000
59.
Yeoman H, Clark DR, DeLuca D: Development of CD4 and CD8 single positive T cells in human thymus organ culture: IL-7 promotes human T cell production by supporting immature T cells.
Dev Comp Immunol
20
:
241
–263,
1996
60.
Hassan J, Reen DJ: IL-7 promotes the survival and maturation but not differentiation of human post-thymic CD4+ T cells.
Eur J Immunol
28
:
3057
–3065,
1998
61.
Li J, Huston G, Swain SL: IL-7 promotes the transition of CD4 effectors to persistent memory cells.
J Exp Med
198
:
1807
–1815,
2003
62.
Fujihashi K, Kawabata S, Hiroi T, Yamamoto M, McGhee JR, Nishikawa S, Kiyono H: Interleukin 2 (IL-2) and interleukin 7 (IL-7) reciprocally induce IL-7 and IL-2 receptors on gamma delta T-cell receptor-positive intraepithelial lymphocytes.
Proc Natl Acad Sci U S A
93
:
3613
–3618,
1996
63.
Kroncke R, Loppnow H, Flad HD, Gerdes J: Human follicular dendritic cells and vascular cells produce interleukin-7: a potential role for interleukin-7 in the germinal center reaction.
Eur J Immunol
26
:
2541
–2544,
1996
64.
Sorg RV, McLellan AD, Hock BD, Fearnley DB, Hart DN: Human dendritic cells express functional interleukin-7.
Immunobiology
198
:
514
–526,
1998
65.
Varas A, Vicente A, Sacedon R, Zapata AG: Interleukin-7 influences the development of thymic dendritic cells.
Blood
92
:
93
–100,
1998
66.
Li L, Masucci MG, Levitsky V: Effect of interleukin-7 on the in vitro development and maturation of monocyte derived human dendritic cells.
Scand J Immunol
51
:
361
–371,
2000
67.
Gombert JM, Herbelin A, Tancrede-Bohin E, Dy M, Carnaud C, Bach JF: Early quantitative and functional deficiency of NK1+-like thymocytes in the NOD mouse.
Eur J Immunol
26
:
2989
–2998,
1996
68.
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 Immunol
161
:
2620
–2628,
1998
69.
Lin CH, Hunig T: Efficient expansion of regulatory T cells in vitro and in vivo with a CD28 superagonist.
Eur J Immunol
33
:
626
–638,
2003
70.
Hoffmann P, Eder R, Kunz-Schughart LA, Andreesen R, Edinger M: Large-scale in vitro expansion of polyclonal human CD4(+)CD25high regulatory T cells.
Blood
104
:
895
–903,
2004
71.
Huber S, Schramm C, Lehr HA, Mann A, Schmitt S, Becker C, Protschka M, Galle PR, Neurath MF, Blessing M: Cutting edge: TGF-beta signaling is required for the in vivo expansion and immunosuppressive capacity of regulatory CD4+CD25+ T cells.
J Immunol
173
:
6526
–6531,
2004
72.
Fu S, Zhang N, Yopp AC, Chen D, Mao M, Zhang H, Ding Y, Bromberg JS: TGF-beta induces Foxp3+ T-regulatory cells from CD4+ CD25− precursors.
Am J Transplant
4
:
1614
–1627,
2004
73.
Tang Q, Henriksen KJ, Bi M, Finger EB, Szot G, Ye J, Masteller EL, McDevitt H, Bonyhadi M, Bluestone JA: In vitro-expanded antigen-specific regulatory T cells suppress autoimmune diabetes.
J Exp Med
199
:
1455
–1465,
2004