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2期 > 正文
编号:11256232
Autoimmune Diabetes Is Suppressed by Transfer of Proinsulin-Encoding Gr-1+ Myeloid Progenitor Cells That Differentiate In Vivo Into Resting
     the Division of Autoimmunity and Transplantation, The Walter and Eliza Hall Institute of Medical Research, Victoria, Australia

    ABSTRACT

    The nature of the T-cell response to antigen is governed by the activation state of the antigen-presenting dendritic cell (DC). Immature or resting DCs have been shown to induce T-cell responses that may protect against the development of autoimmune disease. Effectively harnessing this "tolerogenic" effect of resting DCs requires that it be disease-specific and that activation of DCs by manipulation ex vivo is avoided. We reasoned that this could be achieved by transferring in vivo partially differentiated myeloid progenitor cells encoding a disease-specific autoantigen. With the aim of preventing autoimmune diabetes, we transferred myeloid progenitor cells encoding proinsulin into NOD mice. Bone marrow (BM) was cultured in granulocyte macrophage colony-stimulating factor (GM-CSF) and transforming growth factor-1, a cytokine combination that expands myeloid cells but inhibits terminal DC differentiation, to yield Gr-1+/CD11b+/CD11ceC myeloid progenitor cells and a minor population of CD11c+/CD11b+/CD86lo immature DCs. After transfer, Gr-1+ myeloid cells acquired the characteristics of resting DCs (CD11c+/MHC classIIint/CD86lo/CD40lo). Gr-1+ myeloid cells generated from transgenic NOD mice that expressed proinsulin controlled by a major histocompatibility complex (MHC) class II promoter, but not from wild-type NOD mice, transferred into 4-week-old female NOD mice significantly suppressed diabetes development. The transfer of DC progenitors encoding a disease-specific autoantigen is, therefore, an effective immunotherapeutic strategy that could be applied to humans.

    Antigen-presenting dendritic cells (DCs) determine the functional properties of the T-cells with which they interact. In the absence of activating signals to DCs, such as those mediated through toll-like receptors (1) or CD40 (2), the outcome of DC/T-cell interaction may be a state of T-cell "tolerance." The stage of differentiation, type, and/or location of DCs can determine the form of tolerance that ensues. For example, DCs in lymphoid tissues exist primarily in a "resting" state (3) and present antigen in a manner that appears to result in tolerance due to deletion of antigen-specific T-cells (2,4,5) or induction of unresponsiveness (6), while DCs in mucosal sites induce cytokine skewing and regulatory T-cells (7). Although DCs are promising immunotherapeutic tools, ideally they would target disease-specific T-cells. In the case of autoimmune diseases, this specificity could be achieved by endowing DCs with the ability to express autoantigen. A potential strategy would be to generate cells ex vivo with the phenotypic and functional properties of resting lymphoid tissue DCs, load them with appropriate antigen, and transfer them to effect T-cell tolerance in vivo. This approach is qualified, however, by the plasticity of DCs and the uncertainty of whether they would localize to microanatomic sites for tolerance induction in lymphoid tissues. Activation of DCs can occur during their preparation and manipulation ex vivo (8) or after transfer (9), which may abrogate their tolerogenic potential. This could be avoided by transfering DC progenitor cells that would then differentiate into inherently tolerogenic DCs in vivo. To this end, we generated partially differentiated myeloid cells that retained DC progenitor properties in vitro and in vivo by culture of bone marrow (BM) in granulocyte macrophage colony-stimulating factor (GM-CSF) and transforming growth factor (TGF)-1, a cytokine combination that expands myeloid cells but inhibits terminal DC differentiation. We used the NOD mouse model of spontaneous autoimmune type 1 diabetes, in which proinsulin is a key autoantigen (10,11), to test the ability of DC progenitors encoding proinsulin to suppress the development of autoimmune disease in an antigen-specific manner.

    RESEARCH DESIGN AND METHODS

    Female NODLt/Jax mice were obtained from the Walter and Eliza Hall Institute central breeding facilities. Proinsulin-NOD (PI-NOD) transgenic mice expressing mouse proinsulin II under control of the I-Ek major histocompatibility complex (MHC) class II promoter, described previously (10), were used after breeding to homozygosity. Animals were used in accordance with the Australian Code of Practice for the Care and Use of Animals for Scientific Purposes.

    Reagents, cytokines, and antibodies.

    Culture medium was RPMI-1640 supplemented with 1 mmol/l sodium pyruvate, 0.1 mmol/l nonessential amino acids (both from Gibco, Rockville, MA), 50 eol/l 2-mercaptoethanol (Sigma, St. Louis, MO), and 10% vol/vol FCS (R10) (JRH, Lenexa, KS). Recombinant murine (rm) GM-CSF, interleukin (IL)-4, and tumor necrosis factor (TNF)- were purchased from Peprotech (Rocky Hill, NJ). Recombinant human TGF-1, rmM-CSF, and rmG-CSF were from R&D Systems (Minneapolis, MN). Interferon (IFN)- was kindly provided by Genentech (South San Francisco, CA). Fluorescein isothiocyanate (FITC)-dextran was purchased from Sigma. Antibodies directed against Gr-1 (Ly-6G and RB6-8C5), F4/80 (F4/80), CD11b (M1/70), CD11c (N418), MHC class II (10.2.16 [I-Ak,g7,r,f,s]), MHC class I (M1/42), macrophage colony-stimulating factor receptor (M-CSF R) (AFS-98), CD40 (FGK-45), B220 (RA3-6B2), CD205 (NLDC-145), CD86 (GL-1), and c-kit (ACK-2) were purified from hybridoma supernatants and used as purified mAb or conjugated in house. Streptavidin (SA)-fluorochrome conjugates (SA-FITC, SA-phycoerythrin, SA-allophycocyanin, and SA-Texas red) were from Caltag (Burlingame, CA). mAb directed to CD40 (3/23), MAC-3 (M3/84), CD13 (R3-242), CD62-l (MEL-14), CD31 (MEC13.3), CD43 (S7), CD11a (2D7), and CD49d (R1-2) were purchased from PharMingen (San Diego, CA). Anti-F4/80 was from Caltag (F4/80). Anti-mouse FIRE (6F12) (12) was provided by Dr. Irene Caminschi, Walter and Eliza Hall Institute. 5(6)-Carboxyfluorescein diacetate succinimidyl ester (CFSE) was purchased from Molecular Probes (Eugene, OR).

    BM cultures and cell transfers.

    Mice were euthanized by CO2 asphyxiation and femurs and tibiae removed aseptically. BM was flushed with mouse tonicity PBS/2.5% FCS. Cells were collected by centrifugation and erythrocytes removed by distilled water lysis. After washing in RPMI/10% FCS, cells were plated in 6-well plates (NUNC, Roskilde, Denmark) at 2 x 106/ml in 3 ml R-10 per well. Cultures were initially supplemented with GM-CSF (1 ng/ml) and 2 days later nonadherent cells removed. Remaining adherent cells were then cultured in GM-CSF (1 ng/ml) and TGF-1 (2 ng/ml) for a further 3 days. After a total of 5 days of culture, nonadherent cells were harvested and washed twice in R-10. To generate GM-CSF/IL-4 BMDCs, BM cultures were prepared as described but supplemented with GM-CSF and IL-4 (1 ng/ml of each) for the entire culture period (13). In some experiments, subsets of cells were depleted with either anti-Gr-1 (RB6-8C5) or biotinylated anti-CD11c (N418) and sheep anti-rat Dynabeads or CELLection biotin-binding Dynabeads (Dynal Biotech; Carlton South, Victoria, Australia), respectively, according to the manufacturer’s instructions. Alternatively, CD11c+ cells were depleted with CD11c-phycoerythrin and anti-phycoerythrin magnetic beads (MACS; Miltenyi Biotec, Gladbach, Germany) on a magnetic cell sorter (AutoMACS; Miltenyi Biotec). Unless stated otherwise, to prevent engraftment of hematopoietic stem or progenitor cells that may have remained in the cultures, bulk or depleted cell populations were irradiated (2,000 rads, 60Co source), washed twice in R-10, and resuspended in PBS before injection intravenously into 4-week-old female NOD mice. Irradiation of GM-CSF/TGF-1-generated cells before transfer did not alter their effect on diabetes development. For bulk GM-CSF/TGF-1-cultured BM, 2 x106 cells were transferred intravenously. When cell subsets were depleted, the number of cells transferred was equivalent to that in unseparated bulk GM-CSF/TGF-1-cultured BM, i.e., 1.8 x106 Gr-1+ (CD11c-depleted) cells or 0.2 x 106 CD11c+ (Gr-1-depleted) cells.

    FITC-dextran uptake.

    Quantitation of endocytosis by FITC-dextran uptake was performed as described (13). Briefly, cells were incubated with FITC dextran (1 mg/ml) for 2 h at 37°C or 4°C. Endocytosis was stopped by washing twice with ice-cold PBS containing 2.5% FCS and 0.02% sodium azide, and samples were maintained at 4°C for immunofluorescence staining and flow cytometry.

    Flow cytometric analysis.

    Immunofluorescence staining of cells was performed as described previously (13) for flow cytometry on a FACScan (Becton Dickinson, San Josee, CA). Viable cells were gated on the basis of propidium iodide exclusion. For routine analyses, 1eC2 x 104 live-gated events were collected. For analysis of in vivo DC phenotype, 1eC2 x 106 live-gated events were collected.

    In vitro differentiation assays.

    BM cultured in GM-CSF/TGF-1 was harvested and washed twice, and Gr-1+ or CD11c+ cells were depleted with immunomagnetic beads as described above. After washing, the remaining CD11c+ or Gr-1+ cells were plated in 24-well tissue culture plates (Falcon; Becton Dickinson, Franklin Lakes, NJ) at 106 cells/ml in 1 ml R-10 supplemented with recombinant cytokines (G-CSF, 2 ng/ml; M-CSF, 10 ng/ml; GM-CSF, 5 ng/ml; IL-4, 5 ng/ml; and TNF-, 10 ng/ml) as described in RESULTS. Cultures were maintained in 5% CO2 for up to 7 days.

    In vivo cell tracking and differentiation.

    Cells harvested from GM-CSF/TGF-1-supplemented BM cultures were prepared as described, labeled with 5 eol/l CFSE (14), and injected intravenously. In some experiments, mice were anesthetized with methoxyflurane. Then, the spleen was everted through a keyhole incision and, after being injected with CFSE-labeled cells (5 x 106 in 50 e蘬), gently reinserted into the peritoneal cavity. The wound was closed with surgical clips. Control mice received PBS alone. Mice were killed at defined time points after cell transfer and spleens and other tissues removed. For immunohistology, tissues were embedded in Tissue-Tek OCT freezing medium (Miles, Elkhart, IN). For flow cytometric analysis of in vivo DC development, spleen cell suspensions were prepared using collagenase/EDTA as described (13).

    Adoptive cotransfer assay for regulatory T-cells.

    Adoptive cotransfer assays for antidiabetic regulatory cells were performed as described (15). Spleen cells (2 x 107) from recipients of proinsulin-NOD or wild-type NOD GM-CSF/TGF-1-cultured BM (test cells) and spleen cells (2 x 107) from recently diabetic female NOD mice (diabetogenic cells) were mixed and injected intravenously into NOD.scid mice. Diabetes development was monitored as described below.

    Immunohistology.

    Cryostat sections (5 e) were cut from frozen OCT-embedded tissues, air dried, and fixed with cold 100% ethanol before immunostaining or mounting. Avidin/biotin binding sites were blocked using avidin/biotin blocking reagents (Vector, Burlingame, CA), and nonspecific protein interactions were blocked with 1% BSA. Biotinylated primary antibodies were applied at predetermined optimal concentrations for 1 h at room temperature. After washing, streptavidin horseradish peroxidase (ABC-Elite; Vector) or streptavidin Texas red was applied for a further hour. Immunoperoxidase slides were washed, and staining was developed with enzyme substrate (VectorRed [Vector] or 3,3'-diaminobenzidine, -Fast’ [Sigma]) and counterstained with hematoxylin. Immunofluorescence slides were rinsed and mounted in antifade reagent (DAKO, Carpinteria, CA).

    Cytospins.

    Cytospins were prepared in a cytofuge (Shandon, Pittsburgh, PA) and stained with Diff Quik (Lab Aids, Narrabeen, NSW, Australia) or by immunohistochemistry, as described.

    Assessment of proinsulin production.

    CD11c+ and Gr-1+ cells were purified from day 5 GM-CSF/TGF-1 BM as described. For GM-CSF/IL-4, whole cultures were analyzed. Cells were cultured for 24 h with cytokine (IFN-, 1,000 units/ml, and TNF-, 10 ng/ml) or anti-CD40 (50 e/ml) supplementation. Supernatants were harvested and stored at eC20°C until assayed. Proinsulin production was measured using an insulin enzyme-linked immunosorbent assay kit (Mercodia, Uppsala, Sweden). Proinsulin production was calculated as nanograms of 106 cells per 24 h. The detection limit was <0.1 ng · 106 cellseC1 · 24 heC1.

    Monitoring of diabetes development.

    Mice were tested weekly for urine glucose (Diastix; Bayer, Pymble, NSW, Australia), and if they were glycosuric, they were then tested for blood glucose (Accu-Chek; Roche, Castle Hill, NSW, Australia). Mice were scored diabetic when two consecutive blood glucose readings were >12.0 mmol/l, and then they were killed. Diabetes incidence was plotted as Kaplan-Meier survival curves (GraphPad Prism; GraphPad Software, San Diego, CA).

    Statistical analysis.

    Student’s t test was used for comparison of means (Microsoft Excel). One-way ANOVA with Neuman-Keul’s post test was used for comparisons of multiple groups (GraphPad Prism). Statistical differences in diabetes incidence were analyzed by log-rank test (GraphPad Prism).

    RESULTS

    BM cultured in GM-CSF/TGF-1 contains predominantly partially differentiated Gr-1+ myeloid cells.

    Addition of TGF-1 to GM-CSF-supplemented BM cultures allows myeloid cell expansion but inhibits terminal differentiation of DCs (9,16). Therefore, we surmised that GM-CSF/TGF-1 could be used to generate partially differentiated myeloid progenitors that retain DC development potential. GM-CSF/TGF-1 BM cultures contained a mixture of cell types but were dominated by small round cells with annular or segmented nuclei (Fig. 1A), expressing the myeloid differentiation antigen Gr-1 (Ly-6G; Fig. 1B). These features are characteristic of partially differentiated myeloid cells (17). Florescence-activated cell sorter analysis confirmed the preponderance of Gr-1+ cells and their expression of the myeloid marker CD11b but not the DC-specific marker CD11c (Fig. 2). Gr-1+ cells expressed negligible levels of MHC class I, MHC class II, F4/80, FIRE, MAC 3, CD13 M-CSF R, and c-kit (CD117) (Fig. 2). The adhesion molecules CD49d and CD11a (18,19) used by mature neutrophils to home to sites of inflammation were either not expressed or were expressed at a relatively low level, respectively, by Gr-1+ cells, signifying that these cells were not mature neutrophils. Expression of the myeloid differentiation markers CD43 and CD31 (17,20,21) on Gr-1+ cells was heterogenous, indicating that these cells were present in various differentiation states.

    A small proportion (8.5 ± 2.1% [mean ± SD], n = 20) of GM-CSF/TGF-1 BM cells had a monocyte-like or immature DC-like appearance (Fig. 1C) and expressed low levels of MHC class II restricted primarily to intracellular granules (Fig. 1C). Florescence-activated cell sorter analysis showed that this Gr-1eC fraction comprised almost exclusively cells that expressed low or intermediate levels of CD11c. In addition to MHC class I and moderate levels of MHC class II, CD11c+ cells expressed F4/80, FIRE, MAC 3, CD13, and c-kit (CD117) (Fig. 2A), molecules expressed by immature BMDCs or DCs in vivo. CD11c+ cells also expressed CD62L (L-selectin) used by DC/monocyte precursors to home from blood to lymph nodes (22). The majority of the CD11c+ cells expressed M-CSF R and high levels of CD11b, as described previously for immature BMDCs from NOD mice (13).

    To further define CD11c+ cells from GM-CSF/TGF-1 BM, we compared BM cultured in GM-CSF/TGF-1 and GM-CSF/IL-4, as the latter contains a mix of phenotypically mature and immature DCs (Fig. 1D and online appendix available at http://diabetes.diabetesjournals.org). CD11c+ cells from GM-CSF/TGF-1 BM were phenotypically similar to the immature subset of DCs generated in GM-CSF/IL-4 (online appendix). Endocytic activity, a hallmark of functionally immature (CD11c+/CD86lo) DCs (online appendix), measured in GM-CSF/TGF-1 BM by FITC-dextran uptake, was restricted to CD11c+ cells (online appendix). In contrast to Gr-1+ myeloid cells, CD11c+ cells harvested from GM-CSF/TGF- BM were more differentiated, as evidenced by their rapid development (within 2 days) into DCs in GM-CSF, IL-4, and TNF- or into macrophages in M-CSF or GM-CSF/M-CSF. CD11c+ cells rapidly died in cultures supplemented only with G-CSF (not shown). Collectively, these findings indicate that Gr-1+ cells in GM-CSF/TGF-1 BM are partially differentiated myeloid cells, whereas the minor population of CD11c+ cells are phenotypically and functionally immature DCs.

    Gr-1+ cells are multipotent myeloid progenitors.

    To investigate their differentiation potential, Gr-1+ cells isolated from GM-CSF/TGF-1 BM were exposed to cytokines that drive different myeloid differentiation pathways. In G-CSF, Gr-1+ cells retained their small rounded profile but rapidly acquired highly segmented nuclei and a higher level of Gr-1 expression characteristic of mature granulocytes (Fig. 3). Cells with DC- or macrophage-like characteristics were not detected after 2 days in G-CSF. Consistent with their short lifespan, the numbers of mature granulocytes diminished after 2 days in G-CSF (not shown). In the DC-inducing combination of GM-CSF, IL-4, and TNF-, small numbers of DC-like cells were seen as early as 2 days, and by 7 days large numbers of cells displayed classic DC morphology (Fig. 3). Flow cytometry revealed that exposure to GM-CSF, IL-4, and TNF- resulted in loss of Gr-1 and acquisition of CD11c expression by the majority of cells (Fig. 3). CD11c+ cells were approximately equally distributed between phenotypically mature (MHC class IIhi or CD86hi) and immature (MHC class IIlo or CD86lo) DC subsets (Fig. 3). In M-CSF, Gr-1 expression was lost and adherent macrophage-like cells that retained CD11b expression and acquired low-level expression of the macrophage marker F4/80 appeared.

    Similar phenotype of GM-CSF/TGF-1 BM cells from PI-NOD and wild-type NOD mice.

    No differences were detected between cells generated from PI-NOD and NOD mice in the presence of either GM-CSF/TGF-1 (Fig. 4) or GM-CSF/IL-4 (not shown).

    CD11c+ DCs but not Gr-1+ myeloid cells from PI-NOD transgenic mouse BM produce proinsulin.

    CD11c+ DCs generated from PI-NOD transgenic mouse BM in GM-CSF/IL-4 produced threefold more proinsulin than CD11c+ DCs from GM-CSF/TGF-1 BM (4.2 ± 2.3 vs. 1.3 ± 1.1 ng · 106 cellseC1 · 24 heC1). Cytokines that upregulate MHC class II expression (IFN-/TNF-) enhanced proinsulin production by CD11c+ DCs from GM-CSF/TGF-1 cultures (to 3.4 ± 1.7 ng · 106 cellseC1 · 24 heC1, P < 0.05), whereas agonistic anti-CD40 mAb enhanced proinsulin production by DCs from GM-CSF/IL-4 cultures (to 8.0 ± 2.9 ng · 106 cellseC1 · 24 heC1, P < 0.05) but not GM-CSF/TGF-1 cultures. This is consistent with the expression of CD40 by DCs from GM-CSF/IL-4 BM but not GM-CSF/TGF-1 BM. Purified Gr-1+ cells did not produce detectable proinsulin, even after stimulation with GM-CSF/IFN-/TNF-.

    Diabetes development is suppressed by transfer of proinsulin-encoding Gr-1+ undifferentiated myeloid cells.

    GM-CSF/TGF-1 BM from PI-NOD mice transferred to 4-week-old female NOD mice suppressed diabetes development (Fig. 5A). To identify the cell responsible for this protective effect, CD11c+ or Gr-1+ cells were depleted from GM-CSF/TGF-1 BM before transfer. Depletion of Gr-1+, but not CD11c+, cells abolished the protective effect (Fig. 5B). This was then demonstrated directly and shown to be proinsulin dependent by transfer of purified Gr-1+ cells from GM-CSF/TGF-1 BM of PI-NOD mice (Fig. 5C). Thus, diabetes development was suppressed by transfer of Gr-1+ partially differentiated myeloid cells and not by the CD11c+/CD86lo immature DCs present in GM-CSF/TGF-1 NOD-PI BM.

    Gr-1+ myeloid cells differentiate into DCs in vivo.

    Our hypothesis was that PI-encoding Gr-1+ myeloid cells would protect against diabetes because they differentiated into "tolerogenic" resting DC in vivo. To examine their fate, Gr-1+ myeloid cells purified from PI-NOD GM-CSF/TGF-1 BM were labeled with CFSE to enable visualization after transfer. Two days after intravenous transfer into NOD mice, CFSE-labeled cells (1eC2 per 10x field) could be detected in cryostat sections of spleen, lung, and liver but not in peripheral blood or other tissues (thymus, pancreas, small intestine, kidney, inguinal lymph node, and pancreatic lymph node). CFSE-labeled cells in liver and lung diminished rapidly in number after 2 days but were still detected in spleen (1 per 10x field) at 4 and 6 days posttransfer. Because they were preferentially retained in spleen, we injected CFSE-labeled Gr-1+ cells directly into spleen to examine their differentiation. One day after injection, abundant CFSE-labeled cells with a rounded profile that stained for CD11b and Gr-1, but rarely for MHC class II, were clearly visible. Three days after injection, CFSE-labeled cells were present predominantly in the T-cell areas and had developed a larger, more stellate appearance. Many continued to express CD11b but had lost Gr-1 expression, and 30eC50% had acquired substantial expression of MHC class II and CD11c (Fig. 6). To determine the phenotype of the CD11c+ DCs that developed in vivo, spleens were collected after 3 days for flow cytometry. CD11c+ cells comprised a large proportion of the CFSE-labeled cells in the spleen (Fig. 7A). Three-color flow-cytometry revealed that CFSE+/CD11c+ DCs were almost equally distributed between the CD11c+/CD8+ and CD11c+/CD11b+ subtypes. This contrasted with the dominance of CD11c+/CD11b+ or CD11c+/CD8eC DC subtype (Fig. 7B) normally found in NOD mice (13). Analysis of DEC-205, normally expressed on CD8+ splenic DCs, confirmed the distribution of CFSE+ DCs between CD8+ and CD8eC subtypes (Fig. 7B). CFSE+ DCs derived from Gr-1+ progenitors exhibited a "resting" phenotype, expressing intermediate levels of MHC class II and low levels of the costimulatory/signaling molecules CD86 and CD40, identical to those of unlabeled recipient DCs (Fig. 7C).

    Absence of regulatory, antidiabetogenic cells after transfer of GM-CSF/TGF-1-cultured BM.

    A potential mechanism for the diabetes protective effect of Gr-1+ myeloid cells is induction of regulatory, antidiabetic T-cells. To test for regulatory T-cells, spleen cells from NOD mice that had received NOD or NOD-PI GM-CSF/TGF-1 BM 4 weeks previously were cotransferred with diabetogenic spleen cells from recently diabetic female NOD mice into immunodeficient NOD.scid recipients. The proportion of mice that developed diabetes by 12 weeks after transfer was similar in recipients of spleen cells from mice that received GM-CSF/TGF-1 BM from NOD-PI (78 ± 21%) or wild-type NOD (69 ± 32%) mice.

    DISCUSSION

    Proinsulin-encoding myeloid DC progenitors generated in BM cultured in GM-CSF/TGF-1 differentiate into resting DCs in vivo and suppress the development of autoimmune diabetes in NOD mice. Achieving expression of antigen in resting DCs by transfering antigen-encoding DC progenitors is a novel strategy for preventing autoimmune disease. Previously, the addition of TGF-1 to GM-CSF-supplemented BM cultures was shown to inhibit the final maturation steps of DC development, leading to the generation of phenotypically and functionally immature DCs (9,16). We found that this combination of cytokines also leads to the accumulation of partially differentiated myeloid cells. While Gr-1 is routinely used as a neutrophil marker, it is expressed by a range of myeloid progenitors and, at least transiently, by monocytic cells such as those elicited to the peritoneal cavity (19), as well as by a recently described population of murine blood monocytes that exhibits DC differentiation capacity (23). Our finding that Gr-1+ cells can give rise to DCs and macrophages in vitro is consistent with the observations of others (17,24). In addition, we now show that these cells also differentiate into both major subtypes (CD8+ and CD8eC) of lymphoid tissue DCs in vivo, complementing other evidence (25,26) that myeloid-committed cells are capable of giving rise to both CD8+ and CD8eC DCs in vivo. Undifferentiated myeloid cells (CD11b+/Gr-1+/CD31+) that have been termed natural suppressor cells inhibit CD8+ T-cell activation by antigen-nonspecific nitric oxide-dependent mechanisms (27). A role for these myeloid suppressor cells in our experiments is excluded by their absence of MHC class II expression (27). The crucial requirement for MHC class II+ progeny of Gr-1+ cells for diabetes suppression was demonstrated by the lack of effect of wild-type Gr-1+ cells without the MHC class II-driven proinsulin transgene. Additionally, the low expression of CD31 on Gr-1+ cells generated in GM-CSF/TGF-1 indicates that they are more differentiated than CD31+ myeloid suppressor cells (21,28).

    In contrast to proinsulin-encoding Gr-1+ myeloid cells, proinsulin-encoding immature DCs were not protective. This may be because they were activated in vitro or because exposure to TGF-1 decreases the expression of CCR7 (29), which coordinates DC migration and interaction with T-cells in secondary lymphoid tissues (30). Our findings suggest that Gr-1+ myeloid progenitors may have been overlooked in previous studies examining the tolerance-inducing capacity of GM-CSF/TGF-1-generated immature DCs (9). Because Gr-1+ myeloid cells represent a large proportion of cells in BM (21), they are a potentially important source of DC progenitors that could be harnessed for tolerance induction.

    The mechanism by which proinsulin-encoding myeloid progenitor cells suppress diabetes development remains unclear. Because transferred CFSE-labeled Gr-1+ cells migrated primarily to spleen, liver, and lung, and not thymus, protection from diabetes is likely to be due to peripheral rather than central tolerance. While there is debate about whether specific subsets of DCs, e.g., CD8+ or CD8eC, are "specialized" for peripheral tolerance induction, it is nevertheless clear that DCs must be in a nonactivated or resting state for tolerance to ensue (2,4,5,31). Targeting antigen expression to resting as opposed to activated lymphoid tissue DCs, either by antibody-antigen conjugates or genetically (5), as achieved here for proinsulin, induces deletional tolerance and/or unresponsiveness in antigen-specific T-cells (2,4,6,31). Because both CD8+ and CD8eC DCs that differentiated from transferred Gr-1+ myeloid progenitors exhibited a resting phenotype, we propose that diabetes protection is due to deletion or unresponsiveness of proinsulin-reactive T-cells. This is consistent with our inability to demonstrate the presence of antidiabetogenic Treg. Unfortunately, in common with others (32,33), we are unable to obtain sensitive and reproducible responses to proinsulin by NOD mouse T-cells ex vivo and therefore cannot directly test these possibilities.

    Because of their costimulation dependence (34), CD4+/CD25+ Treg require activation by "mature" DCs to elicit regulatory function (35). Similarly, other forms of Treg in NOD mice may be most efficiently induced by mature DCs (36eC38). This supports suggestions that the impaired maturation potential of NOD DCs (13,39) could lead to reduced Treg activation in vivo (40,41). In this setting, tolerance can be restored by activating DCs in vivo (42) or in an islet antigen-independent manner by adoptively transferring DCs matured ex vivo (36eC38). In contrast, our findings indicate that the ability of resting DCs to induce tolerance is unaffected by the alteration of DC development in NOD mice.

    The literature provides little evidence of the ability of DCs to suppress spontaneously as opposed to experimentally induced autoimmune disease. Attempts to generate tolerogenic DCs that suppress diabetes in an autoantigen-dependent manner after transfer to NOD mice have had little success. In instances where diabetes has been suppressed by DCs generated ex vivo, no requirement for presentation of -cell antigens has been found (36eC38,43eC45). Our findings show that an autoantigen-dependent protective effect, an important requirement for progressing DC-based immunotherapy to the clinic, can be achieved by transferring genetically modified Gr-1+ myeloid progenitors. DC progenitors encoding autoantigen driven by a differentiation stage-specific promoter have several advantages over other DC-based strategies for autoimmune disease prevention. Activation and the risk of antigen presentation that could elicit a pathogenic immune response is minimized. DC progeny express whole autoantigen protein; therefore, assumptions about epitope determinants that are necessary with peptides are avoided, as is the need for substantial quantities of pure protein autoantigen. This immunotherapeutic strategy could be applied to prevent type 1 diabetes and other autoimmune diseases in humans.

    ACKNOWLEDGMENTS

    This work was supported by National Institutes of Healthgrant R21 DK57228, Juvenile Diabetes Research Foundation and Australian National Health and Medical Research Council. The authors thank Dr Irina Caminschi for anti-FIRE antibody and Danielle Cooper and Nicole Clark for expert animal care.

    FOOTNOTES

    Additional information for this article can be found in an online appendix at http://diabetes.diabetesjournals.org.

    BM, bone marrow; CFSE, 5(6)-carboxyfluorescein diacetate succinimidyl ester; DC, dendritic cell; FITC, fluorescein isothiocyanate; GM-CSF, granulocyte macrophage colony-stimulating factor; IFN, interferon; IL, interleukin; M-CSF R, macrophage colony-stimulating factor receptor; MHC, major histocompatibility complex; rm, recombinant murine; SA, streptavidin; TGF, transforming growth factor; TNF, tumor necrosis factor

    REFERENCES

    Kaisho T, Akira S: Regulation of dendritic cell function through Toll-like receptors. Curr Mol Med3 :373 eC385,2003

    Hawiger D, Inaba K, Dorsett Y, Guo M, Mahnke K, Rivera M, Ravetch JV, Steinman RM, Nussenzweig MC: Dendritic cells induce peripheral T-cell unresponsiveness under steady state conditions in vivo. J Exp Med194 :769 eC779,2001

    Wilson NS, El-Sukkari D, Belz GT, Smith CM, Steptoe RJ, Heath WR, Shortman K, Villadangos JA: Most lymphoid organ dendritic cell types are phenotypically and functionally immature. Blood102 :2187 eC2194,2003

    Bonifaz L, Bonnyay D, Mahnke K, Rivera M, Nussenzweig MC, Steinman RM: Efficient targeting of protein antigen to the dendritic cell receptor DEC-205 in the steady state leads to antigen presentation on major histocompatibility complex class I products and peripheral CD8+ T-cell tolerance. J Exp Med196 :1627 eC1638,2002

    Probst HC, Lagnel J, Kollias G, van den Broek M: Inducible transgenic mice reveal resting dendritic cells as potent inducers of CD8+ T-cell tolerance. Immunity18 :713 eC720,2003

    Hawiger D, Masilamani RF, Bettelli E, Kuchroo VK, Nussenzweig MC: Immunological unresponsiveness characterized by increased expression of CD5 on peripheral T-cells induced by dendritic cells in vivo. Immunity20 :695 eC705,2004

    Akbari O, DeKruyff RH, Umetsu DT: Pulmonary dendritic cells producing IL-10 mediate tolerance induced by respiratory exposure to antigen. Nat Immunol2 :725 eC731,2001

    Ridge JP, Di Rosa F, Matzinger P: A conditioned dendritic cell can be a temporal bridge between a CD4+ T-helper and a T-killer cell. Nature393 :474 eC478,1998

    Lu L, Li W, Fu F, Chambers FG, Qian S, Fung JJ, Thomson AW: Blockade of the CD40-CD40 ligand pathway potentiates the capacity of donor-derived dendritic cell progenitors to induce long-term cardiac allograft survival. Transplantation64 :1808 eC1815,1997

    French MB, Allison J, Cram DS, Thomas HE, Dempsey-Collier M, Silva A, Georgiou HM, Kay TW, Harrison LC, Lew AM: Transgenic expression of mouse proinsulin II prevents diabetes in nonobese diabetic mice. Diabetes46 :34 eC39,1997

    Narendran P, Mannering SI, Harrison LC: Proinsulin-a pathogenic autoantigen in type 1 diabetes. Autoimm Rev2 :204 eC210,2003

    Caminschi I, Lucas KM, O’Keeffe MA, Hochrein H, Laabi Y, Kontgen F, Lew AM, Shortman K, Wright MD: Molecular cloning of F4/80-like-receptor, a seven-span membrane protein expressed differentially by dendritic cell and monocyte-macrophage subpopulations. J Immunol167 :3570 eC3576,2001

    Steptoe RJ, Ritchie JM, Harrison LC: Increased generation of dendritic cells from myeloid progenitors in autoimmune-prone non-obese diabetic mice. J Immunol168 :5032 eC5041,2002

    Lyons AB, Parish CR: Determination of lymphocyte division by flow cytometry. J Immunol Methods171 :131 eC137,1994

    Harrison LC, Dempsey-Collier M, Kramer DR, Takahashi K: Aerosol insulin induces regulatory CD8 gamma delta T-cells that prevent murine insulin-dependent diabetes. J Exp Med184 :2167 eC2174,1996

    Yamaguchi Y, Tsumura H, Miwa M, Inaba K: Contrasting effects of TGF-beta 1 and TNF-alpha on the development of dendritic cells from progenitors in mouse bone marrow. Stem Cells15 :144 eC153,1997

    Bierman H, Pietz B, Dreier R, Schnid KW, Sorg C, Sunderkotter C: Murine leukocytes with ring-shaped nuclei include granulocytes, monocytes and their precursors. J Leukoc Biol65 :217 eC231,1999

    Henderson RB, Lim LH, Tessier PA, Gavins FN, Mathies M, Perreti M, Hogg N: The use of lymphocyte function-associated antigen (LFA)-1-deficient mice to determine the role of LFA-1, Mac-1, and alpha4 integrin in the inflammatory response of neutrophils. J Exp Med16 :219 eC226,2001

    Henderson RB, Hobbs JAR, Mathies M, Hogg N: Rapid recruitment of inflammatory monocytes is independent of neutrophil migration. Blood102 :328 eC335,2003

    Hestdal K, Ruscetti FW, Ihle JN, Jacobsen SEW, Dubois CM, Kopp DL, Keller JR: Characterization and regulation of RB6-8C5 antigen expression on murine bone marrow cells. J Immunol147 :22 eC28,1991

    de Bruijn MF, Slieker WA, van der Loo JC, Voerman JS, van Ewijk W, Leenen PJ: Distinct mouse bone marrow macrophage precursors identified by differential expression of ER-MP12 and ER-MP20 antigens. Eur J Immunol24 :2279 eC2284,1994

    Palframan RT, Jung S, Cheng G, Weninger W, Luo Y, Dorf M, Littman DR, Rollins BJ, Zweerink H, Rot A, von Adrian UH: Inflammatory chemokine transport and presentation in HEV: a remote control mechanism for monocyte recruitment to lymph nodes in inflamed tissues. J Exp Med194 :1361 eC1373,2001

    Geissmann F, Jung S, Littman DR: Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity19 :71 eC82,2003

    Oehler L, Majdic O, Pickl WF, Stockl J, Riedl E, Drach J, Rappersberger K, Geissler K, Knapp W: Neutrophil-granulocyte-committed cells can be driven to acquire dendritic cell characteristics. J Exp Med187 :1019 eC1028,1998

    Manz M, Traver D, Miyamoto T, Weissman IL, Akashi K: Dendritic cell potentials of early lymphoid and myeloid progenitors. Blood97 :3333 eC3341,2001

    Gao JX, Liu X, Wen J, Zhang H, Durbin J, Liu Y, Zheng P: Differentiation of monocytic cell clones into CD8 alpha+ dendritic cells (DC) suggests that monocytes can be direct precursors for both CD8 alpha+ and CD8 alpha- DC in the mouse. J Immunol170 :5927 eC5935,2003

    Bronte V, Chappell DB, Apolloni E, Cabrelle A, Wang M, Hwu P, Restifo NP: Unopposed production of granulocyte-macrophage colony-stimulating factor by tumours inhibits CD8+ T-cell responses by dysregulating antigen-presenting cell maturation. J Immunol162 :5728 eC5737,1999

    Nikolic T, de Bruijn MF, Lutz MB, Leenen PJ: Developmental stages of myeloid dendritic cells in mouse bone marrow. Int Immunol15 :515 eC524,2003

    Ogata M, Zhang Y, Wang Y, Itakura M, Zhang YY, Harada A, Hashimoto S, Matsushima K: Chemotactic response toward chemokines and its regulation by transforming growth factor-beta1 of murine bone marrow hematopoietic progenitor cell-derived different subset of dendritic cells. Blood93 :3225 eC3232,1999

    Forster R, Schubel A, Breitfeld D, Kremmer E, Renner-Muller I, Wolf E, Lipp M: CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell99 :23 eC33,1999

    Finkelman FD, Lees A, Birnbaum R, Gause WC, Morris SC: Dendritic cells can present antigen in vivo in either a tolerogenic or immunogenic fashion. J Immunol157 :1406 eC1414,1996

    Kaufman DL, Tisch R, Sarvetnick N, Chatenoud L, Harrison LC, Haskins K, Quinn A, Sercarz E, Singh B, von Herrath M, Wegmann DR, Wen L, Zekzer D: Report from the 1st International NOD Mouse T-Cell Workshop and the follow-up mini workshop. Diabetes50 :2459 eC2463,2001

    Steptoe RJ, Ritchie JM, Harrison LC: Transfer of hematopoietic stem cells encoding autoantigen prevents autoimmune diabetes. J Clin Invest111 :1357 eC1363,2003

    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

    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 Med199 :1467 eC1477,2004

    Feili-Hariri M, Dong X, Alber SM, Watkins SC, Salter RD, Morel PA: Immunotherapy of NOD mice with bone marrow-derived dendritic cells. Diabetes48 :2300 eC2308,1999

    Feili-Hariri M, Falkner DH, Morel PA: Regulatory Th2 response induced following adoptive transfer of dendritic cells in prediabetic NOD mice. Eur J Immunol32 :2021 eC2030,2002

    Morin J, Faideau B, Gagnerault M-C, Lepault F, Boitard C, Boudaly S: Passive transfer of flt-3L-derived dendritic cells delays diabetes development in NOD mice and associates with early production of interleukin (IL)-4 and IL-10 in the spleen of recipient mice. Clin Exp Immunol134 :388 eC395,2003

    Pearson T, Markees TG, Serreze DV, Pierce MA, Marron MP, Wicker LS, Peterson LB, Shultz LD, Mordes JP, Rossini AA, Greiner DL: Genetic disassociation of autoimmunity and resistance to costimulation blockade-induced transplantation tolerance in nonobese diabetic mice. J Immunol171 :185 eC195,2003

    Serreze D, Leiter EH: Defective activation of T suppressor cell function in nonobese diabetic mice: potential relation to cytokine deficiencies. J Immunol140 :3801 eC3807,1988

    Belghith M, Bluestone JA, Barriot S, Megret J, Bach JF, Chatenoud L: TGF-beta-dependent mechanisms mediate restoration of self-tolerance induced by antibodies to CD3 in overt autoimmune diabetes. Nat Med9 :1202 eC1208,2003

    Naumov YN, Bahjat KS, Gausling R, Abraham R, Exley MA, Koezuka Y, Balk SB, Strominger JL, Clare-Salzer M, Wilson SB: Activation of CD1d-restricted T-cells protects NOD mice from developing diabetes by regulating dendritic cell subsets. Proc Natl Acad Sci U S A98 :13838 eC13843,2001

    Papaccio G, Nicoletti F, Pisanti FA, Bendtzen K, Galdieri M: Prevention of spontaneous autoimmune diabetes in NOD mice by transferring in vitro antigen-pulsed syngeneic dendritic cells. Endocrinology141 :1500 eC1505,2000

    Feili-Hariri M, Falkner DH, Gambotto A, Papworth GD, Watkins SC, Robbins PD, Morel PA: Dendritic cells transduced to express interleukin-4 prevent diabetes in nonobese diabetic mice with advanced insulitis. Human Gene Ther14 :13 eC23,2003

    Ma L, Qian S, Liang X, Wang L, Woodward JE, Giannoukakis N, Robbins PD, Bertera S, Trucco M, Fung JJ, Lu L: Prevention of diabetes in NOD mice by administration of dendritic cells deficient in nuclear transcription factor-B activity. Diabetes52 :1976 eC1985,2003(Raymond J. Steptoe, Janin)