当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 糖尿病学杂志 > 2006年 > 第4期 > 正文
编号:11257035
CD4+CD25+ Regulatory T-Cells Inhibit the Islet Innate Immune Response and Promote Islet Engraftment
     1 Department of Gene and Cell Medicine, and Recanati/Miller Transplantation Institute, Mount Sinai School of Medicine, New York, New York

    2 Institute for Cancer Genetics, Columbia University, New York, New York

    3 Division of Nephrology, Mount Sinai School of Medicine, New York, New York

    4 Department of Surgery, Columbia University, New York, New York

    5 Center for Immunobiology, Mount Sinai School of Medicine, New York, New York

    IFN, interferon; IL, interleukin; mAb, monoclonal antibody; PE, phycoethrin; TGF, transforming growth factor; TNF-, tumor necrosis factor-

    ABSTRACT

    Early islet cell loss is a significant problem in clinical islet cell transplantation. Diverse stress stimuli induce innate immune responses in islets that contribute to -cell dysfunction, inflammation, and loss. Here, we show that cytokine-stimulated murine islets express multiple inflammatory chemokines that recruit T-cells and thereby impair islet function in vitro and in vivo. Both nonislet ductal and exocrine elements and the individual islet cellular components contribute to this innate immune response. CD4+CD25+ regulatory T-cells inhibit islet chemokine expression through a cell contacteCdependent, soluble factoreCindependent mechanism and inhibit effector T-cell migration to the islet. Regulatory T-cells can also migrate to stimulated islets. Cotransfer of regulatory T-cells with islets in a transplantation model prevents islet innate immune responses and inflammation and preserves normal architecture and engraftment. Regulatory T-cell inhibition of multiple components of innate immune responses may be a fundamental aspect of their function that influences ischemia-reperfusion injury and adaptive immunity.

    Islet transplantation offers a potential curative treatment for type 1 diabetes. However, in addition to the antialloantigen adaptive response, antigen-independent innate responses initiated during the peritransplant period contribute to islet cell loss and -cell dysfunction. Isolated islets constitutively produce chemokines, and chemokine production is stimulated by local and systemic factors such as brain death, surgical manipulation, ischemia-reperfusion, cytokines, and gene transfer vectors (1eC3). These stimuli impair survival of allogeneic and even syngeneic islet grafts, and decreased survival is associated with increased inflammation in the graft, due in large part to chemokines produced by the islets (4eC7). The role of innate responses in transplant immunity and the benefit of regulating innate immune responses to graft survival have recently been explored (8,9). Modulating innate immunity and chemokine responses may prove to be more effective in preventing detrimental adaptive responses than protocols that rely solely on targeting adaptive immunity. Moreover, because of the redundancy and degeneracy of chemokine and other innate responses, interventions that focus on single molecular pathways are unlikely to successfully control inflammation. Rather, it is important to define mechanisms that simultaneously regulate multiple aspects of the broad innate response.

    CD4+CD25+, anergic, and Foxp3+ regulatory T-cells have emerged as major cellular elements that regulate immune responses (10,11) and have clinical potential to treat diverse immune and inflammatory diseases. Regulatory T-cells have been shown to prevent or even reverse autoimmune diabetes (12) and play important roles in islet alloimmune responses and tolerance to alloantigen (13). In autoimmune diabetes models, regulatory T-cells are able to migrate to the pancreas, the islet, and peripancreatic lymph nodes, showing the importance of chemokines and chemokine responses in their suppressive function (12,14). Regulatory T-cells use a variety of mechanisms to modulate immune responses. In vivo studies suggest that interleukin (IL)-10, CTLA4, and transforming growth factor (TGF)- play important roles in regulatory T-cell effector function (15eC17). In vitro studies show that regulatory T-cells inhibit the activation and proliferation of other T-cells, either directly through a cell contacteCdependent mechanism (18) or indirectly by downregulating antigen-presenting cell activity (19), although regulatory T-cell effector mechanisms remain mostly undefined. Whereas much is known about regulatory T-cell inhibition of adaptive immunity, little is known about their role in other inflammatory events, although recent studies show that regulatory T-cells can inhibit some aspects of innate immunity (20,21). In the current report, we explored the role of regulatory T-cells in modulating islet innate immune responses. The results demonstrate a significant role for regulatory T-cells in modifying many aspects of parenchymal cell innate immunity with subsequent effects on inflammation, islet dysfunction, and graft survival.

    RESEARCH DESIGN AND METHODS

    Inbred BALB/c (H-2d) male mice were purchased from The Jackson Laboratories (Bar Harbor, ME). CXCR3eC/eC mice on a BALB/c background (22) were a gift from Dr. Craig Gerard (Harvard Medical School, Boston, MA). Mice were housed under specific pathogen-free conditions. Animals were treated in strict compliance with regulations and all experiment protocols were approved by the institutional animal care and use committee.

    Antibodies and reagents.

    Recombinant IL-1, tumor necrosis factor- (TNF-), interferon (IFN)-, TGF-2, IL-2, IL-6, IL-10, CCL2/MCP-1, CCL19/ELC, and CXCL9/MIG were purchased from R&D Systems (Minneapolis, MN). CyChromeeCanti-CD4, fluorescein isothiocyanateeCanti-CD25, phycoethrin (PE)eCanti-CD11c, PEeCanti-CD31, anti-CD3, anti-CD4, anti-CD45, antieCLy-6G, antieCF4/80-like receptor, antieCIL-10R1, antieCIL-6, anti-CTLA4, antieCTGF-2, anti-CCL2, anti-CCL5/RANTES, anti-CCL17, and anti-CCL20 were from BD PharMingen (San Diego, CA). Adenovirus vectors expressing -galactosidase under the control of the Rous sarcoma virus promoter (AdRVSLacZ) were grown in our vector core (3,23).

    Cell purification and primary cell culture.

    Single-cell suspensions of murine splenocytes, natural regulatory T-cells (CD4+CD25+), control effector T-cells (CD4+CD25eC), and TGF-eCdriven, de novo regulatory T-cells were purified, generated, and characterized as previously described (24).

    Islet isolation, treatment, and transplantation.

    Islets were isolated from BALB/c mice and transplanted underneath the kidney capsule to streptozotocin-induced diabetic syngeneic mice as previously described (25). Isolated islets were further dispersed into single islet cells by digestion with 0.05 mmol/l trypsin and 0.025 mmol/l EDTA (Sigma-Aldrich, St. Louis, MO) at 37°C for 1 min with pipetting. Islet single cells were then separated into -cells and noneC-cells using -cell autofluorescence (26) with a MoFlo cell sorter. Dendritic cell and endothelial cell depletion was performed with PEeCanti-CD11c and PEeCanti-CD31 staining followed by cell sorting. Islet cultured for 5 days maintained viability and insulin expression (data not shown).

    Groups of 200 islets or 2 x 105 islet single cells were exposed to cytokine mixtures (5 ng/ml TNF-, 100 units/ml IFN-, and 1 ng/ml IL-1) for 1 h in 1 ml serum-free RPMI 1640 culture medium (0.5% BSA, 100 IU/ml penicillin, 0.1 mg/ml streptomycin, and 2 mmol/l L-glutamine) and then washed. Sorted CD4+CD25+ or CD4+CD25eC cells (5 x 104) were added to stimulated islets at a ratio of one lymphocyte to four islet single cells (assuming one islet contains 1,000 single cells). Islets and T-cell populations were cocultured in serum-free RPMI 1640 for 2 days for in vitro experiments or for 2 h followed by cotransplantation to diabetic mice.

    Islet grafts were retrieved from individual animals and embedded in OCT, and consecutive 8-e蘭 sections of the islet grafts were immunostained with anti-CD4, anti-CD45, antieCLy-6G, and antieCF4/80-like receptor. Immunoreactivity was detected using the multilink-HRP ultrasensitive system (BioGenex, San Ramon, CA). Using the color range selection option of Adobe Photoshop, positively stained cells (brown in each case) were selected, and the number of cells per square millimeter in the islet grafts was counted (27). Quantification data present 10 nonoverlapping fields per graft from two individual mice per group.

    RNA isolation, cDNA synthesis, and quantitative real-time PCR.

    RNA isolation, cDNA synthesis, and quantitative real-time PCR were performed as previously described (23,24). The PCR primers were designed to be intron-spanning; sequences are available upon request.

    Multiplex chemokine assay.

    Conditioned islet media were assayed for CCL2, CCL3/MIP-1, CXCL1/KC, CXCL9, and CXCL10/IP-10 with a chemokine 5-Plex bead immunoassay kit (Biosource International, Camarillo, CA), according to the manufacturer’s instructions.

    Migration assays.

    Cells (1eC5 x 105) were added to the top chamber of a 24-well transwell plate, and chemokine-driven migration was quantified as previously described (28). The results are expressed as the percentage of migrated cells. Controls showed that all migration was chemotactic and not chemokinetic (data not shown).

    Statistical analysis.

    Migration and insulin release results represent mean values of duplicate to triplicate samples. Analysis for statistically significant differences was performed with Student’s t test. P < 0.05 was considered a significant difference. Results are expressed as means ± SE.

    RESULTS

    Islets produce chemokines in response to inflammatory cytokines.

    To characterize islet innate immune responses, we identified constitutive and cytokine-stimulated islet chemokines in vitro. Isolated islets were incubated without or with a well-characterized combination of cytokines (TNF-, IL-1, and IFN-) that resemble inflammatory stimuli (1,2,7). After culture for 1 h, islets were washed and cultured for 2 days, and a comprehensive panel of chemokines was screened using quantitative real-time RT-PCR. Figure 1A shows that only some homeostatic chemokines, such as CCL25 and CCL28, have relatively high expression in untreated islets (>5% of ubiquitin). Similar to our previous findings with a cDNA array (7), cytokine stimulation upregulated inflammatory chemokines, such as CCL2, -3, and -5 and CXCL1, -2, -9, -10, and -11 but not homeostatic chemokines. Cytokine-induced upregulation of chemokines was dose dependent (Fig. 1B), and single islet cells produced more than intact islets, probably reflecting the more vigorous chemical and physical manipulation required to make single islet cell suspensions and also more complete exposure of single cells to the cytokine stimuli. These findings were confirmed at the protein level by multiplex chemokine assay of cultured medium (Fig. 1C). Migration assays with conditioned media from cytokine-stimulated islets showed enhanced recruitment of T-cells (Fig. 1D).

    Dendritic cells, endothelial cells, and ductal and exocrine debris are major contributors to islet chemokine responses.

    It is technically inevitable that exocrine and ductal cells will be present as residual debris in isolated islets. These nonislet cells may affect islet chemokine expression. To test this, basal and adenovirus vector-induced cytokine and chemokine expression in islets of 90 (standard islet purity after isolation), 75, and 50% purity (by adding exocrine debris) were assayed by RT-PCR. Adenovirus vector transduction is another innate immune response model efficient in both islet and solid organ tissues (3,7). Islets of 75 and 50% purity express more proinflammatory cytokines (TNF- and inducible nitrogen oxide synthase) and similar chemokines at baseline compared with purer islets (Fig. 2A). Adenovirus vector transduction resulted in greater expression of CCL5 and CXCL10 in islets with debris than in purer islets (Fig. 2A), showing that debris exaggerate the chemokine response synergistically with other inflammatory stimuli.

    Islets contain several cell types: a majority of -cells, a few - and -cells, vascular endothelial cells, and some immune cells (resident macrophages, dendritic cells, and natural killer). NoneC-cells could be separated from single islet cell suspensions by cell sorting using -cell autofluorescence (26). The enriched endocrine -cells express less basal and cytokine-stimulated chemokines than noneC-cells (Fig. 2B). Basal and cytokine-stimulated chemokine expression is significantly decreased after depletion of vascular endothelial cells (CD31+) and dendritic cells (CD11c+) by cell sorting (Fig. 2C). Intact islets were cultured for 5 days to permit intraislet immune cells to migrate out (data not shown) (29). Basal and viral vectoreCstimulated expression of IFN- and inflammatory chemokines (CCL2, CCL5, and CXCL9) is lower after a 5-day culture (Fig. 2D).

    Together, these results demonstrate that different cellular components of islets produce or stimulate chemokines in response to proinflammatory stimuli and suggest that leukocytes, endothelial cells, and ductal and exocrine elements are all major contributors.

    Innate immune responses impair islet function directly or by recruiting effector leukocytes.

    We next characterized the effects of these induced chemokines on islet function. Groups of 150 islets were treated with a mixture of cytokines (TNF-, IFN-, and IL-1) for 1 h, washed, and cultured in a transwell plate, with splenocytes loaded in the top insert and allowed to migrate to the islets for 3 h. After removing the insert, islets were incubated with the migrated splenocytes for another 24 h. Additional groups of isolated islets were incubated with the mixture of cytokines for 1 h, with a mixture of chemokines (CCL2, CCL19, and CXCL9), or directly with splenocytes for 24 h. Islet function was determined by insulin release in response to 5.5 and 20 mmol/l glucose stimulation. None of the experimental treatments altered the viability of the islets, as determined by tetrazolium bromide assay (data not shown), but did reduce both basal insulin release and glucose-stimulated insulin release (Fig. 3). Thus, the cytokine inflammatory stimulus, the innate islet chemokine response, and splenocytes migrating to chemokines all acutely inhibit islet function.

    Regulatory T-cells inhibit islet innate chemokine responses in vitro.

    Because there must be mechanisms that normally regulate islet responses to the many environmental challenges that impinge upon the pancreas, we explored the effect of regulatory T-cells on islet innate immunity by evaluating cytokine-stimulated chemokine induction. Purified islets were treated with cytokines as above and then cocultured for 48 h with natural CD4+CD25+ regulatory T-cells, TGF-eCdriven de novo regulatory T-cells (24), or control CD4+CD25eC T-cells. Fresh or cultured natural CD4+CD25+ regulatory T-cells and TGF-eCdriven de novo regulatory T-cells used in the experiments were routinely tested for Foxp3 expression and suppressive function. Foxp3 mRNA expression was at least 100 (natural regulatory T-cells) to 50 (de novo regulatory T-cells) times higher than in control cells, and regulatory T-cells significantly inhibited anti-CD3 monoclonal antibody (mAb)eCstimulated T-cell proliferation at a ratio of one regulatory T-cell to four T-cells by at least 70%. Islets were handpicked to separate them from the T-cells at the end of the coculture, islet RNA was isolated for RT-PCR, and culture supernatants were saved for protein and migration assays. Fresh or cultured natural CD4+CD25+ regulatory T-cells and TGF-eCdriven de novo regulatory T-cells all inhibit many inflammatory chemokines induced by cytokines, such as CCL2, CCL5, CXCL9, and CXCL10, on the transcriptional (Fig. 4A) and translational (Fig. 4B) levels. Coculture with control T-cells did not have an inhibitory effect on chemokine mRNA expression, protein expression, or migration activity. In migration assays (Fig. 4C), media from cytokine-stimulated islets recruited more splenocytes, and leukocyte recruitment activity was partially reversed by coculture with natural regulatory T-cells or TGF-eCdriven de novo regulatory T-cells. Chemokine induction could be suppressed by natural regulatory T-cells in all experiments, but only CCL2 and CXCL9 of the five tested chemokines were inhibited by TGF-eCdriven de novo regulatory T-cells in some experiments.

    Regulatory T-cells migrate to cytokine-stimulated islets.

    The above results demonstrate that the direct addition of regulatory T-cells to islets inhibits cytokine-stimulated innate chemokine responses. It was next important to determine whether regulatory T-cells can migrate to islets, whether this migration is mechanistically similar to that used by control T-cells, and whether migrated regulatory T-cells have the same inhibitory effect as directly added regulatory T-cells. The chemokine-receptor expression profiles of freshly isolated CD4+CD25+ and CD4+CD25eC T-cells were examined by RT-PCR. Most chemokine receptors are expressed at comparable levels, except that CCR6 expression is higher on CD4+CD25+ regulatory T-cells, whereas CXCR6 expression is higher on CD4+CD25eC cells (Fig. 5A). In migration assays, fresh, cultured, and TGF-eCdriven de novo regulatory T-cells all have similar abilities to migrate to islet-conditioned media, comparable with control fresh CD4+CD25eC T-cells (Fig. 5B). Antibodies to CCL17 (CCR4 ligand) and CCL20 (CCR6 ligand) partially blocked the migration of both control T-cells and regulatory T-cells to cytokine-stimulated islets. Blocking major inflammatory chemokine and receptor pathways by using CXCR3eC/eC T-cells along with anti-CCL2 (CCR2 ligand) and anti-CCL5 (CCR5 ligand) antibodies inhibited CD4+CD25eC control T-cell but not regulatory T-cell migration (Fig. 5C), suggesting that regulatory T-cells use a different but overlapping array of chemokine-receptor interactions to migrate to islets.

    We next tested the suppressive effect of regulatory T-cells that migrated to stimulated islets. Purified islets were treated with cytokines for 1 h and washed, and 5 x 105 fresh regulatory T-cells or control T-cells were placed in the upper chamber of the transwell system and allowed to migrate to the stimulated islets overnight. Approximately 10% of the input cells, i.e., 5 x 104 cells, migrated to the islets, equivalent to the number of T-cells placed directly into the islet cultures. After migration, islets were cultured with the migrated T-cells for an additional 24 h. The migrated regulatory T-cells inhibit cytokine-stimulated inflammatory chemokine expression in islets as potently as directly applied regulatory T-cells (Fig. 5D).

    Regulatory T-cells inhibit islet chemokine responses through a cell contacteCdependent, soluble factoreCindependent mechanism.

    Although most investigators have found that the suppressive properties of CD4+CD25+ regulatory T-cells in vitro are strictly cell contact dependent and independent of soluble agents, studies in vivo suggest that IL-10 and TGF- are important for their suppressive effects (15eC17). To determine whether regulatory T-cells suppress chemokine responses by direct contact or soluble mediators in the islet cultures, a 0.4-e蘭 transwell system was used to prevent direct contact of regulatory T-cells and islets during coculture. The results in Fig. 6A show that direct contact of regulatory T-cells and cytokine-stimulated islets is required to inhibit chemokine expression. To determine whether soluble mediators might also contribute to regulatory T-cell activity, antieCTGF-, anti-IL-10, anti-IL-10R1, and anti-CTLA-4 mAbs were added to isleteCregulatory T-cell cocultures separately or as a mixture. The results in Fig. 6B and C demonstrate that these antibodies do not abrogate regulatory T-cell inhibition of cytokine-stimulated chemokine induction in islets, suggesting these molecules play a limited role in regulatory T-cell function in this model.

    Regulatory T-cells inhibit IL-6 production by stimulated islets.

    In addition to expressing inflammatory chemokines in response to innate immune stimulation, islets are also able to express a variety of other molecules. In particular, we evaluated IL-6, another acute inflammatory cytokine expressed by islets (30) that has potent chemotactic activities (31). The results in Fig. 7A show that cytokine-stimulated islets express IL-6 mRNA, and IL-6 is inhibited by coculture with regulatory T-cells. The inhibitory effect of regulatory T-cells on IL-6 expression is cell contact dependent and IL-10, CTLA4, and TGF- independent (Fig. 7B). Transwell migration assays showed that IL-6 is able to recruit splenocytes (Fig. 7C). Thus, regulatory T-cells modulate islet innate immunity by suppressing both chemokines and cytokines, limiting their effect on effector leukocyte recruitment. Additional experiments showed that IL-6 is a better chemoattractant for CD4+CD25eC T-cells than for CD4+CD25+ regulatory T-cells (Fig. 7D). This along with the finding that blockade of inflammatory chemokine-receptor pathways inhibit effector T-cell but not regulatory T-cell migration (Fig. 5C) suggests that regulatory T-cells may preferentially inhibit the recruitment of effector leukocytes but not the influx of additional regulatory T-cells.

    Regulatory T-cells protect islet grafts from innate immune injury and enhance islet graft function.

    We next explored the utility of regulatory T-cells in an islet transplant model to determine whether these cells could regulate leukocyte infiltration and protect grafts at early stages after transplantation. To focus on innate immunity, we used a marginal mass, syngeneic islet transplantation model in BALB/c mice (3). As shown in Fig. 8A, untreated control but not cytokine-stimulated islets reverse hyperglycemia 24 h after transplantation. Cotransfer of regulatory T-cells but not control T-cells with the islets re-establishes diabetes cure in cytokine-stimulated islets.

    Islet grafts were harvested 1 and 4 days after transplantation and assayed for chemokine expression by RT-PCR and inflammatory infiltrates by immunohistochemistry. Inflammatory CCL5 and CXCL10 expression predominates at day 1 and decreases by day 4 in controls, confirming that this group of chemokines contributes to the very early, innate response. Cytokine-stimulated islets express higher CXCL10 and similar CCL5 levels as controls. Their cognate receptors, CCR5 and CXCR3, known to be expressed on activated Th1 cells (32), are expressed at higher levels in cytokine-treated islets on both days 1 and 4, suggesting recruitment of more inflammatory cells in these islets. Transplanted control islets express higher levels of CCL5 and CXCL10, compared with control islets in vitro (Fig. 8B vs. Fig. 1A), suggesting that surgical manipulation and ischemia induced inflammatory chemokine expression in islets. Cotransfer of regulatory T-cells with cytokine-treated islet grafts reversed inflammatory chemokine and chemokine-receptor expression to levels even lower than those in control islets, suggesting that regulatory T-cells protected islets not only from cytokine stimulation but also from other stimuli such as surgical manipulation and ischemia (Fig. 8B). Histological examination showed a significant leukocytic infiltrate and islet destruction in cytokine-stimulated grafts. The infiltrate was more intense on day 1 compared with day 4, commensurate with RT-PCR data. Immunohistochemistry identified that neutrophils were the predominant CD45+ leukocytes infiltrating cytokine-stimulated islet grafts, along with a few macrophages and CD4 T-cells that were also present (Fig. 8CeCE; data not shown). In grafts with cotransferred regulatory T-cells, there was significantly less inflammatory infiltrate with preservation of islet structure. Because there were few islet-associated lymphocytes in the regulatory T-cell group, this suggests that the cells may have apoptosed or migrated to draining lymph nodes.

    DISCUSSION

    In this study, we characterized the islet innate immune response using a cytokine-stimulated model that resembles the inflammatory events that impinge on donor islets and explored the potential role of regulatory T-cells in regulating innate immunity and facilitating islet engraftment. We found that islets respond to multiple stimuli, which act through cytokine receptors, toll-like receptors, and probably other receptor pathways, by producing inflammatory chemokines. These chemokines attract lymphocytes, amplify inflammation, impair islet engraftment, and decrease graft survival. These chemokines are produced both by ductal and exocrine contaminants and by multiple different cellular components of islets, including -cells, dendritic cells, and vascular endothelial cells. Regulatory T-cells are able to migrate to inflamed islets and inhibit islet chemokine expression and production, thereby decreasing inflammation and improving graft survival.

    Recent studies demonstrate that suppressive properties of regulatory T-cells are not limited to effects on T-cell responses but also include inhibition of pathology mediated by the cells of innate immune system (20,21,33). In our study, regulatory T-cells directly interact with pancreatic islets, in the absence of effector T-cells, to inhibit the innate response of islets for chemokine production. These findings may partly explain the current failure to fully define regulatory T-cell effector function because most investigations have focused only on regulatory T-cell interactions with effector T-cells and antigen-presenting cell in adaptive immunity. The results here suggest direct interactions between regulatory T-cells and parenchymal cells, so investigations of regulatory T-cell mechanisms will now have to focus on interactions that can be sustained by parenchymal cells. Given recent considerations that adaptive immunity is highly dependent on innate immunity (34), the results here suggest that manipulation of regulatory T-cells to modulate innate immunity will not only decrease acute injury but also decrease innate amplification of subsequent adaptive immunity.

    As a functional unit, islets are composed of multiple cellular components, although few studies have explored the individual contribution of the different components to the islet innate immune response. In the current study, we demonstrated that dendritic cells, vascular endothelial cells, and ductal and exocrine debris contribute to a large degree to islet chemokine production and the innate immune response (Fig. 2). Because islet revascularization is critical to islet engraftment and survival (25,35) and because dendritic cells can function both as initiators of the immune response and as inducers of peripheral tolerance (36), it will be necessary to modulate islet endothelial cell and dendritic cell function rather than simply deplete these cells as strategies to improve islet transplantation. This may be achieved via the introduction of regulatory T-cells into islets or modification of dendritic cells or vascular endothelial cells to support the migration, expansion, or differentiation of regulatory T-cells.

    The ability of regulatory T-cells to inhibit a range of immune responses and target a variety of cells suggests that they use multiple different effector mechanisms. Many in vivo studies show that IL-10, CTLA4, and TGF- play important roles in regulatory T-cell function (15eC17). There is evidence that PD-1/PD-L1 (37), CD30 (38), TRANCE-RANK (39), cytolytic activity through granzyme A (40) or granzyme B (41), Lag-3 (42), and production of indolemine 2,3-dioxygenase by dendritic cells and HO-1 by regulatory T-cells (43) are also involved in regulatory T-celleCsuppressive function. In the current study, we found that the suppressive function of regulatory T-cells on islets is cell contact dependent and IL-10, TGF-, or CTLA4 independent (similar observations rev. in 11). Our findings suggest investigations of regulatory T-celleCparenchymal cell, eCdendritic cell, or eCvascular endothelial cell interactions as novel avenues of experimental inquiry. Because parenchymal and vascular endothelial cells would not be expected to express the ligands for most of the mechanisms cited above, it is likely that there are other important regulatory T-cell molecular mediators remaining to be characterized.

    As for other effector T-cells, regulatory T-cells must migrate to the proper location, traffic to a microdomain within that location, adhere to stromal cells, and deploy effector functions. Although a number of studies have examined regulatory T-cell chemotaxis, migration, and receptor expression, there is little agreement among the reports. Regulatory T-cells have been variously described by different groups as bearing exclusively CCR4 and CCR8 (44) or higher CCR6 (45). We found that most chemokine receptors are expressed at comparable levels on both CD4+CD25+ and CD4+CD25eC cells (Fig. 5A). However, similar chemokine-receptor expression profiles do not necessarily mean identical receptor usage for chemotaxis. In the current study, we found that regulatory T-cells and CD4+CD25eC T-cells migrate similarly to mixed islet chemokines but not to IL-6, blockade of CCR4 and CCR6 partially inhibited the migration of both control T-cells and regulatory T-cells, and yet blockade of CCL2, CCL5, and CXCR3 inhibited only control T-cells. Together, the findings suggest that regulatory T-cells and effector T-cells use an overlapping array of chemokine-receptor interactions for migration and trafficking. Thus, depending on the nature of the inflammatory stimuli, different populations of regulatory or effector T-cells may traffic to a region of tissue injury. Regulatory T-cells can also change receptor or integrin expression patterns to home to lymphoid organs or exit to inflamed tissue (46). As demonstrated here, regulatory T-cells migrate to islets, interact with islet parenchymal cells or intraislet leukocytes, and suppress islet immune responses. Regulatory T-cells can respond to CCL19 and CCL21 (47,48) and migrate to peripancreatic lymph nodes (14). In the current study, cotransferred regulatory T-cells prevent islet graft inflammation, but they were not present in large numbers under the kidney capsule, even 1 day after transfer, suggesting that they may have had an initial effect on the islets, and then migrated to draining lymph nodes or even efferent lymphatics and the general circulation to have more widespread regional or systemic effects on inflammation. Our current studies are directed to determine whether cotransferred regulatory T-cells do migrate to draining lymph nodes to inhibit effector cell recruitment.

    ACKNOWLEDGMENTS

    J.S.B. has received National Institutes of Health Grants R01-AI-42840, R01-AI-44929, and R01-AI-62765 and Juvenile Diabetes Research Foundation Grant 1-2005-16. S.A.L. has received National Institutes of Health Grant R01-DK-067381.

    We gratefully acknowledge Drs. Yaozhong Ding and Jordi Ochando for critical review and discussion of the work.

    FOOTNOTES

    D.C. and N.Z. contributed equally to this work.

    The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

    REFERENCES

    Frigerio S, Junt T, Lu B, Gerard C, Zumsteg U, Hollander GA, Piali L: Beta cells are responsible for CXCR3-mediated T-cell infiltration in insulitis. Nat Med 8:1414eC1420, 2002

    Bottino R, Balamurugan AN, Tse H, Thirunavukkarasu C, Ge X, Profozich J, Milton M, Ziegenfuss A, Trucco M, Piganelli JD: Response of human islets to isolation stress and the effect of antioxidant treatment. Diabetes 53:2559eC2268, 2004

    Zhang N, Schrppel B, Chen D, Fu S, Hudkins KL, Zhang H, Murphy B, Sung RS, Bromberg JS: Adenovirus transduction induces expression of multiple chemokines and chemokine receptors in murine beta cells and pancreatic islets. Am J Transplant 3:1230eC1241, 2003

    Lee I, Wang L, Wells AD, Ye Q, Han R, Dorf ME, Kuziel WA, Rollins BJ, Chen L, Hancock WW: Blocking the monocyte chemoattractant protein-1/CCR2 chemokine pathway induces permanent survival of islet allografts through a programmed death-1 ligand-1-dependent mechanism. J Immunol 171:6929eC6935, 2003

    Schrppel B, Zhang N, Chen P, Zang W, Chen D, Hudkins KL, Kuziel WA, Sung R, Bromberg JS, Murphy B: Differential expression of chemokines and chemokine receptors in murine islet allografts: the role of CCR2 and CCR5 signaling pathways. J Am Soc Nephrol 15:1853eC1861, 2004

    Abdi R, Means TK, Ito T, Smith RN, Najafian N, Jurewicz M, Tchipachvili V, Charo I, Auchincloss H Jr, Sayegh MH, Luster AD: Differential role of CCR2 in islet and heart allograft rejection: tissue specificity of chemokine/chemokine receptor function in vivo. J Immunol 172:767eC775, 2004

    Schrppel B, Zhang N, Chen P, Chen D, Bromberg JS, Murphy B: Role of donor-derived monocyte chemoattractant protein-1 in murine islet transplantation. J Am Soc Nephrol 16:444eC451, 2005

    El-Sawy T, Miura M, Fairchild R: Early T cell response to allografts occurring prior to alloantigen priming up-regulates innate-mediated inflammation and graft necrosis. Am J Pathol 165:147eC157, 2004

    Goldstein DR, Tesar BM, Akira S, Lakkis FG: Critical role of the Toll-like receptor signal adaptor protein MyD88 in acute allograft rejection. J Clin Invest 111:1571eC1578, 2003

    Sakaguchi S, Sakaguchi N, Shimizu J, Yamazaki S, Sakihama T, Itoh M, Kuniyasu Y, Nomura T, Toda M, Takahashi T: Immunologic tolerance maintained by CD25+ CD4+ regulatory T cells: their common role in controlling autoimmunity, tumor immunity, and transplantation tolerance. Immunol Rev 182:18eC32, 2001

    Shevach EM: CD4+ CD25+ suppressor T cells: more questions than answers (Review). Nat Rev Immunol 2:389eC400, 2002

    Bour-Jordan H, Salomon BL, Thompson HL, Szot GL, Bernhard MR, Bluestone JA: Costimulation controls diabetes by altering the balance of pathogenic and regulatory T cells. J Clin Invest 114:979eC987, 2004

    Davies JD, O’Connor E, Hall D, Krahl T, Trotter J, Sarvetnick N: CD4+ CD45RB low-density cells from untreated mice prevent acute allograft rejection. J Immunol 163:5353eC5357, 1999

    Green EA, Gorelik L, McGregor CM, Tran EH, Flavell RA: CD4+CD25+ T regulatory cells control anti-islet CD8+ T cells through TGF-beta-TGF-beta receptor interactions in type 1 diabetes. Proc Natl Acad Sci U S A 100:10878eC10883, 2003

    Read S, Malmstrom V, Powrie F: Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25+CD4+ regulatory cells that control intestinal inflammation. J Exp Med 192:295eC302, 2000

    Marie JC, Letterio JJ, Gavin M, Rudensky AY: TGF-beta1 maintains suppressor function and Foxp3 expression in CD4+CD25+ regulatory T cells. J Exp Med 201:1061eC1067, 2005

    Kingsley CI, Karim M, Bushell AR, Wood KJ: CD25+CD4+ regulatory T cells prevent graft rejection: CTLA-4- and IL-10-dependent immunoregulation of alloresponses. J Immunol 168:1080eC1086, 2002

    Thornton AM, Shevach EM: CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. J Exp Med 188:287eC296, 1998

    Vendetti S, Chai JG, Dyson J, Simpson E, Lombardi G, Lechler R: Anergic T cells inhibit the antigen-presenting function of dendritic cells. J Immunol 165:1175eC1181, 2000

    Maloy KJ, Salaun L, Cahill R, Dougan G, Saunders NJ, Powrie F: CD4+CD25+ T(R) cells suppress innate immune pathology through cytokine-dependent mechanisms. J Exp Med 197:111eC119, 2003

    Murphy TJ, Choileain NN, Zang Y, Mannick JA, Lederer JA: CD4+CD25+ regulatory T cells control innate immune reactivity after injury. J Immunol 174:2957eC2963, 2005

    Hancock WW, Lu B, Gao W, Csizmadia V, Faia K, King JA, Smiley ST, Ling M, Gerard NP, Gerard C: Requirement of the chemokine receptor CXCR3 for acute allograft rejection. J Exp Med 192:1515eC1520, 2000

    Chen D, Ding Y, Zhang N, Schrppel B, Fu S, Zang W, Zhang H, Hancock WW, Bromberg JS: Viral IL-10 gene transfer inhibits the expression of multiple chemokine and chemokine receptor genes induced by inflammatory or adaptive immune stimuli. Am J Transplant 3:1538eC1549, 2003

    Fu S, Zhang N, Yopp AC, Chen D, Mao M, Chen D, Zhang H, Ding Y, Bromberg JS: TGF-beta induces Foxp3 + T-regulatory cells from CD4+CD25eC precursors. Am J Transplant 4:1614eC1627, 2004

    Zhang N, Richter A, Suriawinata J, Harbaran S, Altomonte J, Cong L, Zhang H, Song K, Meseck M, Bromberg J, Dong H: Elevated vascular endothelial growth factor production in islets improves islet graft vascularization. Diabetes 53:963eC970, 2004

    Pipeleers D, in’t Veld PA, Van de Winkel M, Maes E, Schuit FC, Gepts W: A new in vitro model for the study of pancreatic A and B cells. Endocrinology 117:806eC816, 1985

    Saiki T, Ezaki T, Ogawa M, Matruno K: Trafficking of host- and donor-derived dendritic cells in rat cardiac transplantation: allosensitization in the spleen and hepatic nodes. Transplantation 71:1806eC1815, 2001

    Honig SM, Fu S, Mao X, Bromberg JS: FTY720 stimulates multidrug transporter- and cysteinyl leukotriene-dependent T cell chemotaxis to lymph nodes. J Clin Invest 111:627eC637, 2003

    Markmann JF, Tomaszewski J, Posselt AM, Levy MM, Woehrle M, Barker CF, Naji A: The effect of islet cell culture in vitro at 24 degrees C on graft survival and MHC antigen expression. Transplantation 49:272eC277, 1990

    Ozasa T, Newton NR, Dallman MJ, Shimizu S, Gray DW, Morris PJ: Cytokine gene expression in pancreatic islet grafts in the rat. Transplantation 64:1152eC1159, 1997

    Weissenbach M, Clahsen T, Weber C, Spitzer D, Wirth D, Vestweber D, Heinrich PC, Schaper F: Interleukin-6 is a direct mediator of T cell migration. Eur J Immunol 34:2895eC2906, 2004

    Lohmann T, Laue S, Nietzschmann U, Kapellen TM, Lehmann I, Schroeder S, Paschke R, Kiess W: Reduced expression of Th1-associated chemokine receptors on peripheral blood lymphocytes at diagnosis of type 1 diabetes. Diabetes 51:2474eC2480, 2002

    Taams LS, van Amelsfort JM, Tiemessen MM, Jacobs KM, de Jong EC, Akbar AN, Bijlsma JW, Lafeber FP: Modulation of monocyte/macrophage function by human CD4+CD25+ regulatory T cells. Hum Immunol 66:222eC230, 2005

    Germain RN: An innately interesting decade of research in immunology. Nat Med 10:1307eC1320, 2004

    Brissova M, Fowler M, Wiebe P, Shostak A, Shiota M, Radhika A, Lin PC, Gannon M, Powers AC: Intraislet endothelial cells contribute to revascularization of transplanted pancreatic islet. Diabetes 53:1318eC1325, 2004

    Banchereau J, Palucka AK: Dendritic cells as therapeutic vaccines against cancer. Nat Rev Immunol 5:296eC306, 2005

    Baecher-Allan C, Brown JA, Freeman GJ, Hafler DA: CD4+CD25 high regulatory cells in human peripheral blood. J Immunol 167:1245eC1253, 2001

    Dai Z, Li Q, Wang Y, Gao G, Diggs LS, Tellides G, Lakkis FG: CD4+CD25+ regulatory T cells suppress allograft rejection mediated by memory CD8+ T cells via a CD30-dependent mechanism. J Clin Invest 113:310eC317, 2004

    Green EA, Choi Y, Flavell RA: Pancreatic lymph node-derived CD4(+)CD25(+) Treg cells: highly potent regulators of diabetes that require TRANCE-RANK signals. Immunity 16:183eC191, 2002

    Grossman WJ, Verbsky JW, Barchet W, Colonna M, Atkinson JP, Ley TJ: Human T regulatory cells can use the perforin pathway to cause autologous target cell death. Immunity 21:589eC601, 2004

    Gondek DC, Lu LF, Quezada SA, Sakaguchi S, Noelle RJ: Cutting edge: contact-mediated suppression by CD4+CD25+ regulatory cells involves a granzyme B-dependent, perforin-independent mechanism. J Immunol 174:1783eC1786, 2005

    Huang CT, Workman CJ, Flies D, Pan X, Marson AL, Zhou G, Hipkiss EL, Ravi S, Kowalski J, Levitsk HI, Powell JD, Pardoll DM, Drake CG, Vignali DA: Role of LAG-3 in regulatory T cells. Immunity 21:503eC513, 2004

    Brusko TM, Wasserfall CH, Agarwal A, Kapturczak MH, Atkinson MA: An integral role for heme oxygenase-1 and carbon monoxide in maintaining peripheral tolerance by CD4+CD25+ regulatory T cells. J Immunol 174:5181eC5186, 2005

    Iellem A, Mariani M, Lang R, Recalde H, Panina-Bordignon P, Sinigaglia F, D’Ambrosio D: Unique chemotactic response profile and specific expression of chemokine receptors CCR4 and CCR8 by CD4(+)CD25(+) regulatory T cells. J Exp Med 194:847eC853, 2001

    Kleinewietfeld M, Puentes F, Borsellino G, Battistini L, Rotzschke O, Falk K: CCR6 expression defines regulatory effector/memory-like cells within the CD25(+)CD4+ T-cell subset. Blood 105:2877eC2886, 2005

    Huehn J, Siegmund K, Lehmann JC, Siewert C, Haubold U, Feuerer M, Debes GF, Lauber J, Frey O, Przybylski GK, Niesner U, de la Rosa M, Schmidt CA, Brauer R, Buer J, Scheffold A, Hamann A: Developmental stage, phenotype, and migration distinguish naive- and effector/memory-like CD4+ regulatory T cells. J Exp Med 199:303eC313, 2004

    Gavin MA, Clarke SR, Negrou E, Gallegos A, Rudensky A: Homeostasis and anergy of CD4(+)CD25(+) suppressor T cells in vivo. Nat Immunol 3:33eC41, 2003

    Fu S, Yopp AC, Mao X, Chen D, Zhang N, Chen D, Mao M, Ding Y, Bromberg JS: CD4+ CD25+ CD62+ T-regulatory cell subset has optimal suppressive and proliferative potential. Am J Transplant 4:65eC78, 2004(Dongmei Chen, Nan Zhang, )