Abstract
Type 1 diabetes is an autoimmune disease characterized by destruction of the pancreatic islet beta cells that is mediated primarily by T cells specific for beta cell antigens. Insulin administration prolongs the life of affected individuals, but often fails to prevent the serious complications that decrease quality of life and result in significant morbidity and mortality. Thus, new strategies for the prevention and treatment of this disease are warranted. Given the important role of dendritic cells (DCs) in the establishment of peripheral T cell tolerance, DC-based strategies are a rational and exciting avenue of exploration. DCs employ a diverse arsenal to maintain tolerance, including the induction of T cell deletion or anergy and the generation and expansion of regulatory T cell populations. Here we review DC-based immunotherapeutic approaches to type 1 diabetes, most of which have been employed in non-obese diabetic (NOD) mice or other murine models of the disease. These strategies include administration of in vitro-generated DCs, deliberate exposure of DCs to antigens before transfer and the targeting of antigens to DCs in vivo. Although remarkable results have often been obtained in these model systems, the challenge now is to translate DC-based immunotherapeutic strategies to humans, while at the same time minimizing the potential for global immunosuppression or exacerbation of autoimmune responses. In this review, we have devoted considerable attention to antigen-specific DC-based approaches, as results from murine models suggest that they have the potential to result in regulatory T cell populations capable of both preventing and reversing type 1 diabetes.
Keywords: dendritic cells, diabetes, immunotherapy
Type 1 diabetes
Type 1 diabetes is an organ-specific autoimmune disease characterized by progressive loss of the insulin-producing beta cells that reside within the pancreatic islets [1]. This loss is primarily T cell-mediated and orchestrated by CD4+ and CD8+ T cells specific for beta cell antigens [2]. Present in both type 1 diabetes patients and in non-obese diabetic (NOD) mice, a well-studied model of the disease, these T cells employ a variety of mechanisms to bring about beta cell elimination [3]. These include Fas/FasL interactions and perforin- and cytokine-mediated cell killing. Although systemic pharmacological immunosuppression can halt the autoimmune attack [4], its side effects render it unacceptable for routine use in type 1 diabetes patients. Insulin injections prolong life but are often unable to prevent the serious diabetic complications that are associated with significant morbidity and mortality. Thus, there is an ongoing worldwide effort to develop new strategies for the prevention and treatment of this disease.
Nearly two decades ago, Clare-Salzler and colleagues reported that dendritic cells (DCs) isolated from the pancreatic lymph nodes of NOD mice could prevent diabetes development when transferred adoptively to young recipients [5]. These findings spurred efforts to develop DC-based interventions for type 1 diabetes. The overall favourable safety profile of DC-based therapies revealed by cancer immunotherapy trials has provided further inspiration for such work [6–15]. Here we will discuss the progress that has been made in the area of DC-based therapeutics for type 1 diabetes, with a special emphasis on antigen-specific approaches. We will limit our discussion to ‘conventional’ DCs, as the therapeutic promise of plasmacytoid DCs in type 1 diabetes has been reviewed recently [16].
An introduction to DCs
The identification of DCs was reported by Steinman and Cohn in 1973 [17], a discovery that was driven by a desire to ‘understand immunogenicity’[18]. One of the initial demonstrations of the immunogenic role of DCs was the finding that isolated murine lymphoid organ DCs were potent stimulators of the mixed leucocyte reaction [19]. However, two decades later, when an antigen was delivered specifically to a subset of murine DCs in vivo (i.e. those expressing the endocytic receptor DEC-205), the predicted outcome of a robust immune response did not occur [20]. Antigen-specific tolerance was observed instead, as cognate T cells were largely deleted or rendered unresponsive. It is now understood that in the steady state (i.e. in the absence of infection), DCs are largely immature and present antigens to T cells in a tolerogenic manner, an activity that is important for the establishment of peripheral tolerance [21]. Such DCs are characterized by low expression of CD40 and the T cell co-stimulatory molecules CD80 and CD86. In contrast, in the case of host exposure to a pathogen, DCs undergo a maturation process, e.g. in response to microbial-derived products, that leads to increased antigen presentation and expression of T cell co-stimulatory molecules and T cell responses of a type appropriate to combat the offending pathogen [22]. It should be noted that CD4+CD25+ regulatory T cells (Tregs) are best expanded, at least in vitro, when CD86high DCs are used as antigen-presenting cells [23,24]. Thus, it is important to keep in mind that a certain level of DC maturity may be important for the generation of Tregs capable of inhibiting autoimmune disease [25].
The development of conventional lymphoid organ DCs in mice has been clarified recently [26]. The macrophage and DC precursor gives rise to the common DC precursor (the source of both conventional and plasmacytoid DCs). The next developmental stage for the conventional lymphoid organ DC is the pre-DC. The pre-DCs expand in the bone marrow and differentiate to conventional DCs within the spleen and lymph nodes, where they proliferate in response to Flt3L [27]. A number of DC subsets have been described phenotypically in both mice and humans [28]. Some of these are known to be functionally specialized [29]. For example, in mice, the DC subset expressing CD8 and DEC-205 is specialized for capture of dying cells [30] and cross-presentation of antigens on class I major histocompatibility complex (MHC) molecules [31–33], while CD8-DCIR2+ DCs are proficient at presentation of peptides on class II MHC [32].
Strategies adopted by DCs to induce peripheral T cell tolerance
In addition to their well-established role in central tolerance [34], DCs employ a variety of diverse strategies and pathways to maintain T cell tolerance in the periphery (Fig. 1). Apart from induction of deletional tolerance of peripheral T cells [20,35], DCs in the steady state can also render them anergized [20] as a result of antigen recognition without sufficient co-stimulation [36]. T cell co-inhibitory molecules that transduce negative signals, such as cytotoxic T lymphocyte antigen-4 (CTLA-4) [37] or programmed death-1 (PD-1) [38,39], also participate in these processes. For example, steady-state DCs utilize both the PD-1 and CTLA-4 pathways to induce peripheral tolerance of CD8+ T cells [40]. In addition to induction of deletion or anergy, DCs can induce increased expression of CD5 on activated T cells that leads to hyporesponsiveness, at least in the setting of the induced autoimmune disease, experimental acute encephalomyelitis [41]. Expression of Fas on antigen-presenting cells is also important for the maintenance of peripheral tolerance and the avoidance of autoimmunity [42], while the production of indoleamine 2,3-dioxygenase (IDO) by DCs is involved in peripheral tolerance in certain specialized settings [43,44]. Finally, DCs are involved in the in vivo expansion of thymic-derived natural CD4+CD25+ Tregs[45] as well as the induction of adaptive forkhead box P3 (FoxP3+) Tregs[45–48] and CD8+ Tregs[49], and interleukin (IL)-7 produced by immature DCs appears to function as a CD4+CD25+ Treg survival factor [50].
Fig. 1.

Different strategies of induction of peripheral tolerance by dendritic cells (DCs). Steady-state DCs can induce deletion or anergy of cognate autoreactive T cells either through lack of co-stimulation or by recruitment of co-inhibitory molecules. Another efficient method of tolerance induction is the generation and expansion of antigen-specific regulatory T cells. Apart from deletion of DCs by Fas expression, DCs can induce tolerance directly in the periphery by production of indoleamine 2,3-dioxygenase.
The role of DCs during the natural history of type 1 diabetes
Multiple lines of investigation indicate that priming of pathogenic beta cell-specific T cells occurs in the pancreatic lymph nodes. For example, adoptive transfer of 5,6-carboxy-succinimidyl-fluorescein-ester (CFSE)-labelled transgenic CD4+ BDC2.5 T cells (specific for a beta cell antigen) revealed proliferation of the transferred cells selectively in the pancreatic lymph nodes and before the onset of insulitis [51]. Removal of the pancreatic lymph nodes of 3-week-old NOD mice prevented diabetes development [52], again suggesting that autoreactive T cell priming occurs at this site. While DCs are responsible for this presentation of beta cell antigens [53–55], it is important to realize that the outcome of this can be T cell deletion or regulation instead of pathogenic T cell priming [53,54], even in the diabetes-prone NOD mouse [56]. Serreze and colleagues found that a significant proportion of transferred islet-reactive CD8+ AI4 T cells underwent apoptosis in the pancreatic lymph nodes of NOD mice, but not in other sites such as the mesenteric lymph nodes [56]. In addition, pancreatic lymph node-residing AI4 T cells were less responsive to antigen when compared to cells isolated from the mesenteric lymph nodes [56]. These observations are consistent with the finding that transfer of pancreatic lymph node DCs to young (4-week-old) NOD mice could prevent diabetes development [5]. Such results serve as the foundation for current efforts to explore the immunotherapeutic potential of DCs in type 1 diabetes.
DC-based therapeutic strategies that do not incorporate islet antigens
Morel's group showed that DCs generated from the bone marrow of NOD mice by culture in granulocyte–macrophage colony-stimulating factor (GM-CSF), IL-4 and fetal bovine serum (FBS) could prevent diabetes in some recipients when administered as 3-weekly intravenous injections to young (5-week-old) NOD mice [57]. These bone marrow-derived DCs (BMDCs) expressed class II MHC, CD80, CD86 and CD40 in vitro, although CD40 expression was subsequently diminished upon in vivo administration. Pulsing of the DCs with a mixture of defined beta cell peptides [heat shock protein 60 (HSP60437–460), glutamic acid decarboxylase 65 (GAD65509–528) and GAD65524–543] before transfer did not augment their ability to prevent disease. Mice receiving DCs (pulsed with beta cell peptides or not) exhibited an increased immunoglobulin G1 (IgG1) response to GAD65509–528. As IL-4 facilitates class-switching to this isotype, the investigators speculated, and showed later [58], that DC administration leads to the stimulation of regulatory T helper type 2 (Th2) T cell responses, as determined by cytokine production in response to anti-T cell receptor (anti-TCR) stimulation.
Subsequent to these studies, von Herrath demonstrated that murine BMDCs generated in FBS caused systemic immune deviation in recipients due to a Th2 cell response to FBS-derived proteins [59]. This resulted in impaired clearance of a lymphocytic choriomeningitis virus (LCMV) infection, which normally relies on a Th1 response and interferon (IFN)-γ-producing cytotoxic CD8+ T cells. This important study urged investigators to avoid DC exposure to FBS in their preclinical studies, in order to more effectively mimic future clinical trials where FBS would not be used. It also demonstrated the need to monitor possible systemic immunosuppressive effects of DC administration that might have undesirable clinical consequences, e.g. impaired viral clearance.
Genetically modified DCs have also been employed in preclinical models of type 1 diabetes. BMDCs transduced with a lentiviral vector encoding IL-4 were able to prevent disease in old (12-week-old) NOD recipients, i.e. well after the onset of insulitis, whereas unmodified DCs could not [60]. BMDCs engineered to express galectin-1 by transduction with a recombinant adenovirus were capable of delaying the onset of diabetes induced in immunodeficient NOD recipients by transfer of splenocytes from diabetic NOD females [61]. This is consistent with the recent finding that stimuli that induce tolerogenic DCs, such as IL-10 and 1,25-dihydroxyvitamin D3, also increase their expression of galectin-1 [62].
In addition to viral vectors, treatment with anti-sense oligonucleotides has been used to engineer DCs having a tolerogenic phenotype. Giannoukakis and Trucco used anti-sense oligonucleotides targeting the CD40, CD80 and CD86 messages to treat BMDCs from NOD mice in order to engineer phenotypically immature DCs [63]. When these DCs were administered intraperitoneally to 5–8-week-old NOD mice, a single injection was able to prolong the time to diabetes onset. The therapeutic effect correlated with an increased percentage of splenic CD4+CD25+ (presumably regulatory) T cells. Systemic immunosuppression was not observed, as splenocytes from DC-treated mice were able to respond to alloantigens in vitro. These investigators showed subsequently that four weekly injections of anti-sense oligonucleotide-treated DCs, beginning at 8 weeks of age, resulted in prevention of disease in all recipients [50].
BMDCs from NOD mice have also been manipulated by treatment with decoy double-stranded oligonucleotides containing nuclear factor-kappa B (NF-κB) binding sites [64]. The treated DCs exhibited reduced NF-κB activity and suppression of co-stimulatory molecule expression and IL-12 production. When administered as a single intravenous injection to NOD mice at 6–7 weeks of age, NF-κB-deficient DCs had a dramatic disease-preventive effect, while untreated DCs or those treated with control oligonucleotides were only modestly beneficial.
When contemplating therapeutic administration of DCs, it is important to consider the in vivo trafficking patterns of the administered cells. Creusot and Fathman showed that BMDCs administered intraperitoneally to mice accumulated preferentially in the pancreatic lymph nodes as opposed to other lymph nodes or the spleen [65]. This was the case even in non-diabetes-prone mouse strains. This could explain why intraperitoneal administration of anti-sense oligonucleotide-treated DCs delayed diabetes onset but did not result in systemic immunosuppression [63]. Intravenously administered BMDCs accumulated predominantly in the spleen and the pancreatic and tracheal lymph nodes [60,65]. This finding is consistent with the speculation [57] that intravenously administered DCs can acquire islet antigens in vivo (a process that would take place in the pancreatic lymph nodes) and, thus, can modulate effector and regulatory T cell responses to diabetes-relevant antigens even without deliberate prior antigen treatment.
Exposure of therapeutic DCs to antigen ex vivo
The original observation that DCs from the pancreatic lymph nodes could prevent diabetes when transferred to NOD mice, while those from other sites could not, suggested the potential importance of the incorporation of beta cell antigens into DC-based therapeutics for this disease [5]. As reviewed recently [66], a variety of immunosuppressive and anti-inflammatory compounds, e.g. vitamin D3 and mycophenolate mofetil, can endow DCs with a tolerogenic functional phenotype. Cytokines such as IL-10 can behave similarly [67]. This suggests a therapeutic strategy for type 1 diabetes in which tolerogenic DCs would be generated in vitro and then exposed to beta cell antigens prior to administration. Such an approach was employed recently by the von Herrath group [59], who utilized the rat insulin promoter (RIP)-LCMV model of type 1 diabetes in which disease is induced upon LCMV infection. BMDCs were generated in the presence of GM-CSF, IL-10 and normal mouse serum, and then pulsed with a viral peptide recognized by CD8+ T cells. When the pulsed DCs were administered intraperitoneally to mice 10 and 3 days prior to LCMV infection, only 45% of the animals developed diabetes, whereas 80% of those treated with unpulsed DCs became diabetic. A reduced expansion of viral-specific T cells in response to viral infection was also observed in mice treated with peptide-pulsed DCs. This study supports the idea that ex vivo-generated tolerogenic DCs, when exposed to disease-relevant antigens, can deliver therapeutic benefit in type 1 diabetes.
Targeting of antigens to DCs in vivo
In a recent thoughtful review of DC-based immunotherapeutic strategies for human diseases, the disadvantages of ex vivo antigen loading of DCs were discussed [68]. These include a requirement for leukapheresis, the inability to manipulate DCs within their natural milieu and a requirement for a tailor-made ‘product’ for each patient, resulting in labour-intensive procedures and high costs. It is for reasons such as these that we [69] and others [70] are exploring the utility of in vivo delivery of beta cell antigens to DCs in the prevention and treatment of type 1 diabetes.
DEC-205: an endocytic receptor involved in antigen processing and presentation
DCs employ a variety of molecules, such as the Fc receptors, the macrophage mannose receptor (MMR) and DEC-205 [71], to execute receptor-mediated endocytosis of antigens. Of these, DEC-205 (Ly75/CD205) has the special ability to uptake and subsequently present antigen via both class I [35] and class II MHC pathways [72]. DEC-205 is a type 1 transmembrane protein homologous to MMR and phospholipase A2 [71]. Being a member of this family, this 205 kDa protein has a characteristic short cytoplasmic domain that mediates adsorptive endocytosis. It is made of a single polypeptide chain and contains 10 lectin domains, which is a deviation from the eight lectin domains of the MMR family. Named from the first observations of its abundant expression on DCs and thymic epithelial cells in mice using the rat monoclonal antibody non-lymphoid dendritic cells (NLDC)-145 [73,74], DEC-205 has a more diverse distribution. B cells from various sources such as spleen, lymph node and peritoneal exudates express DEC-205, but at a much lower level than on DCs [75]. Immunohistochemical staining showed expression of DEC-205 on the follicular B cells, bone marrow stroma and pulmonary airway epithelium [76].
Although found predominantly on DCs, DEC-205 is not expressed ubiquitously on all DC subsets. In the mouse thymus, all DC show DEC-205 expression, the majority of which are CD8+[77]. In contrast, murine spleen shows three subsets of DC: CD4+CD8-DEC205-CD11b+, CD4-CD8-DEC205-CD11b+ and CD4-CD8+DEC205+CD11b-[77]. Two additional populations can be traced in the lymph nodes, which show lower expression of CD8 but high to moderate expression of DEC-205 [78]. Moreover, non-lymphoid DCs such as the Langerhans cells of the skin and also BMDCs generated in the presence of GM-CSF show high expression of DEC-205 [75,79]. While humans do not have a DC subset that is CD8+, most DCs in the T cell areas of human spleen and lymph nodes co-express CD11c and DEC-205 [80,81].
DC-induced tolerance upon delivery of antigen via DEC-205
In a pioneering study, Hawiger and colleagues fused an immunogenic peptide from hen egg lysozyme (HEL) to the carboxyl terminus of the heavy chain of NLDC-145 [20]. They injected mice with the hybrid antibody and found that the anti-DEC-205/HEL could deliver antigen to DCs leading to CD4+ T cell activation and proliferation. However, further investigation showed that this treatment led ultimately to the deletion of many of the antigen-specific T cells and that the remaining T cells were unresponsive and could not mount an immune response to subsequent challenge with HEL administered with complete Freund's adjuvant (CFA), showing the induction of HEL-specific tolerance. However, when the same treatment was performed in conjunction with an agonistic anti-CD40 antibody, the outcome was prolonged T cell activation and immunity. Thus, it could be inferred that DCs in the steady state, i.e. in the absence of additional stimuli, act as inducers of antigen-specific peripheral tolerance.
Subsequently, ovalbumin (OVA) was coupled chemically to anti-DEC-205 and was found to permit DCs to present a cognate peptide to OVA-specific CD8+ T cells [35]. The antibody-mediated delivery was much more efficient than administration of soluble OVA alone. In agreement with the earlier study that utilized anti-DEC-205/HEL and CD4+ T cells [20], when anti-DEC-205/OVA was targeted to DCs in the steady state in vivo there was an initial burst of proliferation of the OVA-specific CD8+ T cells which was followed by their deletion. The treated mice also showed tolerance to subsequent rechallenge with OVA along with CFA, which was, however, nullified when anti-CD40 was co-administered with the anti-DEC-205/OVA reagent. Therefore, the DEC-205 receptor can deliver antigen to DCs for presentation to both CD4+ and CD8+ T cells, and when that is performed in the steady state it leads to deletional tolerance or anergy of the antigen-specific T cells.
Targeting steady-state immature DCs with antigen-linked anti-DEC-205 antibody, apart from inducing anergy and deletion of cognate T cells [20,35], can also lead to the induction and/or expansion of Tregs[47,82]. Anti-DEC-205/OVA drove short-lived proliferation of OVA-specific CD4+ T cells in vivo and led to the induction of CD25+/CTLA-4+ T cells with regulatory properties which could suppress proliferation and IL-2 production of conventional CD4+ T cells in a dose-dependent manner [82]. This phenomenon was corroborated further in CD4+ and CD8+ T cell-driven hypersensitivity models, where suppression of immune responses could be achieved in vivo by the induction of CD4+CD25+ Tregs by antigen-linked anti-DEC-205. To investigate further whether DCs are able to induce Tregs from truly naive FoxP3- CD4+ T cells, peptide ligands were targeted to DCs through DEC-205 and FoxP3 expression was analysed at the single-cell level [47]. In this study, which used T cells from Rag2−/− TCR transgenic mice to exclude pre-existing FoxP3+ cells, it was shown that the converted Tregs expressed FoxP3 just as do natural Tregs. It was also demonstrated that minute antigen doses with suboptimal DC activation were necessary for Treg induction, which was enhanced further by the addition of transforming growth factor (TGF)-β or in the absence of IL-2. Importantly, these FoxP3+ Tregs could be expanded by immunogenic presentation of antigen and also retained their surface phenotype and suppressor activity.
Recently, Yamazaki and Steinman reported that CD8+DEC205+ splenic DCs are particularly well equipped to induce FoxP3+ Tregs from FoxP3- precursors [45]. This occurs in the presence of low doses of antigen and requires TGF-β expressed by the DEC-205+ DCs themselves. This may explain partially why, in some cases, DC targeting by antigen-linked anti-DEC-205 antibody led to the conversion of conventional CD4+ T cells to CD25+CD4+ Tregs[47,82].
DEC-205-mediated antigen delivery: application to type 1 diabetes
The therapeutic potential of DEC-205-mediated antigen delivery has begun to be explored in mouse models of type 1 diabetes [69,70]. The first such study utilized a CD4+ T cell-driven model in which mice express haemagglutinin (HA) under the control of the rat insulin promoter (INS) and an I-Ed-restricted TCR specific for HA110–120. These mice have a diabetes incidence of 40%. When treated with HA peptide-linked anti-DEC-205 repeatedly from birth until 12–16 weeks of age, diabetes was prevented in most animals. While extensive deletion of the HA-specific T cells was not observed, they exhibited increased expression of FoxP3, CTLA-4, TGF-β and IL-10, which the authors presumed was responsible for the disease-protective effect of anti-DEC-205/HA.
In addition to CD4+ T cells, the involvement of cytotoxic CD8+ T cells in the pathogenesis of type 1 diabetes is well established in NOD mice [83]. Furthermore, deletion of a single CD8+ T cell specificity by soluble peptide therapy has shown some therapeutic benefit in this model [84,85]. Therefore, beta cell antigenic epitopes targeted by CD8+ T cells are potential candidates for antigen-based tolerogenic strategies. Keeping this in mind, in our laboratory a superagonist mimotope peptide recognized by the AI4 CD8+ T cell clone was delivered to DCs in NOD mice using peptide-linked anti-DEC-205 [69]. Transferred antigen-specific T cells were found to undergo initial proliferation, only to be deleted later. When the treated mice were rechallenged with the mimotope, along with CFA, no immune response could be induced, indicative of antigen-specific tolerance. These findings demonstrated that targeting of DCs with a beta cell antigen, even in the context of the ongoing autoimmune activity present in NOD mice, could lead to deletion of autoreactive CD8+ T cells and subsequent tolerance induction.
The wide variety of antigens and T cell epitopes targeted in type 1 diabetes in both NOD mice and humans [2] suggests that simple deletion of a single antigenic specificity, or even several, may be unable to provide durable clinical benefit. However, we believe that targeting of antigens to DEC-205+ DCs holds promise due to its additional potential to facilitate the expansion and/or induction of Tregs[45,47,70,82]. The importance of FoxP3+ Tregs in type 1 diabetes is demonstrated by the fact that children with a congenital defect in FoxP3 expression rapidly develop a variety of autoimmune diseases, including type 1 diabetes [86,87]. CD4+CD25+ Tregs have also been shown to prevent or reverse diabetes in NOD mice [23,88–90]. Importantly, DCs from NOD mice were found to be capable of expanding CD4+CD25+ BDC2.5 T cells in vitro[23]. These islet-specific Tregs were a potent inhibitor of diabetes development in NOD mice, even though multiple antigenic specificities participate in beta cell demise in this model [2]. These DC-expanded islet-specific Tregs, when administered to NOD mice, could also block diabetes long after the initiation of insulitis and caused long-lasting reversal of hyperglycaemia even after development of overt disease [90].
Antigen choice
When developing DEC-205-mediated therapeutic strategies for type 1 diabetes, the choice of antigen is not a straightforward one. As mentioned, multiple antigens are targeted by T cells in both NOD mice and type 1 diabetes patients [2]. Particularly in humans, it is unclear which of these are the most ‘important’, i.e. critical for disease initiation and/or progression. However, accumulating evidence suggests that insulin is a key target of pathogenic T cells in type 1 diabetes in both NOD mice and humans and that it may even be the long-sought-after ‘initiating antigen’ in this disease [91–103]. In NOD mice, establishment of tolerance to insulin can lead to prevention of diabetes [95,100,101] as well as remission of established disease [93]. Importantly, CD8+ and CD4+ T cell responses to insulin have also been reported in type 1 diabetes patients [91,94,96,99,102]. Furthermore, in humans, the non-MHC locus that confers the strongest susceptibility to type 1 diabetes is the insulin gene variable number of tandem repeats (VNTR) regulatory region [104], and disease-associated alleles are correlated with reduced thymic expression of the insulin gene [105]. We are exploring the feasibility of DEC-205-mediated delivery of the entire preproinsulin molecule, rather than only the known epitopes targeted by effector T cells. This strategy should facilitate translation to patients expressing diverse MHC molecules. In addition, the epitopes recognized by insulin-specific regulatory T cells are largely uncharacterized and could differ from those targeted by pathogenic effector T cells [106]. The finding that DC-expanded Tregs of a single specificity can both prevent and reverse type 1 diabetes in NOD mice [23,90] provides critical support for this approach.
Insuring against harm
We found that peptide-linked anti-DEC-205 could induce tolerance even in NOD mice with ongoing islet inflammation [69]. However, when contemplating the translation of such a strategy to humans, there is a concern that antigen delivery to DCs in the context of an inflammatory environment could lead to exacerbation of a pathogenic autoimmune response rather than tolerance induction. One potential remedy to be considered is the simultaneous use of siRNA specific for co-stimulatory molecules which could be targeted to the DCs in vivo through either DEC-205 or another DC receptor. In vivo siRNA delivery, although difficult to achieve, has been conducted through cell surface receptors by other groups [107–110]. Another possible strategy would be to use microsphere carriers of anti-sense oligonucleotides that can down-modulate co-stimulatory molecules on DCs in vivo[111].
Considerations of disease stage
DC-based therapeutics for type 1 diabetes should be considered at all stages of the disease, including prediabetes, new-onset diabetes and the setting of islet transplantation. In general, it has been easier to prevent diabetes in the NOD mouse model than it has been to reverse it [112]. For this and other reasons, it has been argued that prevention should be the goal [106]. However, given the more favourable risk to benefit ratio represented by new-onset diabetes patients, it may be easier to conduct clinical trials in such individuals, and there are examples of successful reversal of type 1 diabetes in NOD mice (e.g. by transfer of DC-expanded Tregs[90] or in vivo delivery of anti-sense oligonucleotides for CD80, CD86 and CD40 [111]). Studies in animal models suggest that DC-based immunotherapeutic strategies might also be utilized to facilitate islet transplant tolerance [113–115].
DC-based therapies in humans
DC-based therapeutic approaches designed to stimulate immune responses to tumours have been employed in patients with advanced cancers for nearly 15 years, with one of the earliest reports appearing in 1996 [10]. Such studies utilize DCs pulsed with tumour antigens [10], tumour antigen-derived peptides [6,7,11,12,15] or tumour lysates [9], or DCs transfected with tumour antigen cDNA (e.g. Muc1) [13], total tumour RNA [14] or RNA encoding tumour antigens (e.g. prostate-specific antigen) [8]. As reviewed, such therapies are safe, and tumour regression has been observed in some patients [22]. Multiple studies have revealed that mature DCs are optimal for stimulation of anti-tumour immune responses [7,11]. In contrast, and of clear relevance for type 1 diabetes therapeutics, when immature DCs pulsed with an influenza matrix peptide were administered to healthy controls [49,116] the outcome was inhibition of the function of peptide-specific effector CD8+ T cells and the appearance of peptide-specific IL-10-producing CD8+ T cells [116], as well as regulatory CD8+ T cells that required cell–cell contact to exert their suppressive effects [49].
At this time, the use of DCs in humans is being extended slowly beyond cancer immunotherapy to treatment of infectious diseases [117] and autoimmune diseases including type 1 diabetes [118] and rheumatoid arthritis [119]. As discussed in an earlier section of this review, the administration of DCs rendered phenotypically immature by treatment with anti-sense oligonucleotides for CD80, CD86 and CD40 can prevent diabetes development in NOD mice [50,63]. The safety of this strategy is currently being evaluated in a Phase I clinical trial of long-standing adult type 1 diabetes patients in which autologous DCs are being generated from blood precursors after leukapheresis and treated with anti-sense oligonucleotides in vitro[118]. In this study, which began in 2007, the DCs are injected intradermally at a site proximal to the pancreas where they are expected to migrate to the nearest lymph nodes, including those of the pancreas. This same group reported that in vivo administration of microspheres incorporating the anti-sense oligonucleotides is capable of preventing and reversing type 1 diabetes development in NOD mice [111], and they anticipate human trials in the near future [118]. If approved, this strategy would greatly simplify the therapeutic protocol, as it would eliminate the need for leukapheresis and in vitro DC generation and treatment with oligonucleotides.
Conclusion
Despite the DC defects that have been reported in NOD mice [120–123], a variety of DC-based immunotherapeutic strategies have shown great promise in this model, as we have summarized here (Fig. 2). Now the challenge will be to translate these approaches to patients. The ongoing investigation of the safety of phenotypically immature autologous DCs administered to type 1 diabetes patients represents a giant step forward in this regard [118]. In our view, efforts should be made to minimize the possibility of global immunosuppression by utilizing strategies that confer antigen specificity, either by deliberate incorporation of beta cell antigens or by exploiting the migration patterns of administered DCs [60,65]. Continued evaluation of such strategies, particularly in humanized models of the disease [124], should help to allay translational fears and facilitate the transit of DC-based therapies to patients.
Fig. 2.

Dendritic cell (DC)-based therapeutic strategies in the fight against diabetes. DCs have shown promising results in the treatment of type 1 diabetes when used alone or in conjunction with various other molecules (for details, see text). Targeting of DCs with islet antigens has also proved successful. However, most of these studies have been performed in non-obese diabetic (NOD) mice or other mouse models for type 1 diabetes. Although they serve as excellent models, they show significant differences from the disease in humans. Therefore, the ultimate goal, as well as challenge, is to translate these successful studies to human patients. It is hoped that the ongoing Phase I clinical trial with autologous immature DCs (iDC) (see Ref. [118]) will establish the safety of DC administration in type 1 diabetes patients.
Acknowledgments
We apologize to our colleagues whose work could not be cited individually due to space restrictions. Relevant research by our group is supported by the National Institutes of Health, the Juvenile Diabetes Research Foundation International, the American Diabetes Association and the Irma T. Hirschl/Monique Weill-Caulier Trust.
Disclosure
The authors declare no conflicts of interest.
References
- 1.Gianani R, Eisenbarth GS. The stages of type 1A diabetes: 2005. Immunol Rev. 2005;204:232–49. doi: 10.1111/j.0105-2896.2005.00248.x. [DOI] [PubMed] [Google Scholar]
- 2.DiLorenzo TP, Peakman M, Roep BO. Translational mini-review series on type 1 diabetes: systematic analysis of T cell epitopes in autoimmune diabetes. Clin Exp Immunol. 2007;148:1–16. doi: 10.1111/j.1365-2249.2006.03244.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Mathis D, Vence L, Benoist C. β-Cell death during progression to diabetes. Nature. 2001;414:792–8. doi: 10.1038/414792a. [DOI] [PubMed] [Google Scholar]
- 4.Feutren G, Papoz L, Assan R, et al. Cyclosporin increases the rate and length of remissions in insulin-dependent diabetes of recent onset. Results of a multicentre double-blind trial. Lancet. 1986;2:119–24. doi: 10.1016/s0140-6736(86)91943-4. [DOI] [PubMed] [Google Scholar]
- 5.Clare-Salzler MJ, Brooks J, Chai A, Van Herle K, Anderson C. Prevention of diabetes in nonobese diabetic mice by dendritic cell transfer. J Clin Invest. 1992;90:741–8. doi: 10.1172/JCI115946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Banchereau J, Palucka AK, Dhodapkar M, et al. Immune and clinical responses in patients with metastatic melanoma to CD34+ progenitor-derived dendritic cell vaccine. Cancer Res. 2001;61:6451–8. [PubMed] [Google Scholar]
- 7.de Vries IJ, Lesterhuis WJ, Scharenborg NM, et al. Maturation of dendritic cells is a prerequisite for inducing immune responses in advanced melanoma patients. Clin Cancer Res. 2003;9:5091–100. [PubMed] [Google Scholar]
- 8.Heiser A, Coleman D, Dannull J, et al. Autologous dendritic cells transfected with prostate-specific antigen RNA stimulate CTL responses against metastatic prostate tumors. J Clin Invest. 2002;109:409–17. doi: 10.1172/JCI14364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Holtl L, Zelle-Rieser C, Gander H, et al. Immunotherapy of metastatic renal cell carcinoma with tumor lysate-pulsed autologous dendritic cells. Clin Cancer Res. 2002;8:3369–76. [PubMed] [Google Scholar]
- 10.Hsu FJ, Benike C, Fagnoni F, et al. Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells. Nat Med. 1996;2:52–8. doi: 10.1038/nm0196-52. [DOI] [PubMed] [Google Scholar]
- 11.Jonuleit H, Giesecke-Tuettenberg A, Tuting T, et al. A comparison of two types of dendritic cell as adjuvants for the induction of melanoma-specific T-cell responses in humans following intranodal injection. Int J Cancer. 2001;93:243–51. doi: 10.1002/ijc.1323. [DOI] [PubMed] [Google Scholar]
- 12.Mackensen A, Herbst B, Chen JL, et al. Phase I study in melanoma patients of a vaccine with peptide-pulsed dendritic cells generated in vitro from CD34+ hematopoietic progenitor cells. Int J Cancer. 2000;86:385–92. doi: 10.1002/(sici)1097-0215(20000501)86:3<385::aid-ijc13>3.0.co;2-t. [DOI] [PubMed] [Google Scholar]
- 13.Pecher G, Haring A, Kaiser L, Thiel E. Mucin gene (MUC1) transfected dendritic cells as vaccine: results of a phase I/II clinical trial. Cancer Immunol Immunother. 2002;51:669–73. doi: 10.1007/s00262-002-0317-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Su Z, Dannull J, Heiser A, et al. Immunological and clinical responses in metastatic renal cancer patients vaccinated with tumor RNA-transfected dendritic cells. Cancer Res. 2003;63:2127–33. [PubMed] [Google Scholar]
- 15.Thurner B, Haendle I, Roder C, et al. Vaccination with mage-3A1 peptide-pulsed mature, monocyte-derived dendritic cells expands specific cytotoxic T cells and induces regression of some metastases in advanced stage IV melanoma. J Exp Med. 1999;190:1669–78. doi: 10.1084/jem.190.11.1669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Nikolic T, Welzen-Coppens JM, Leenen PJ, Drexhage HA, Versnel MA. Plasmacytoid dendritic cells in autoimmune diabetes – potential tools for immunotherapy. Immunobiology. 2009;214:791–9. doi: 10.1016/j.imbio.2009.06.002. [DOI] [PubMed] [Google Scholar]
- 17.Steinman RM, Cohn ZA. Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution. J Exp Med. 1973;137:1142–62. doi: 10.1084/jem.137.5.1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Steinman RM. Dendritic cells: understanding immunogenicity. Eur J Immunol. 2007;37(Suppl 1):S53–60. doi: 10.1002/eji.200737400. [DOI] [PubMed] [Google Scholar]
- 19.Steinman RM, Witmer MD. Lymphoid dendritic cells are potent stimulators of the primary mixed leukocyte reaction in mice. Proc Natl Acad Sci USA. 1978;75:5132–6. doi: 10.1073/pnas.75.10.5132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hawiger D, Inaba K, Dorsett Y, et al. Dendritic cells induce peripheral T cell unresponsiveness under steady state conditions in vivo. J Exp Med. 2001;194:769–79. doi: 10.1084/jem.194.6.769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic cells. Annu Rev Immunol. 2003;21:685–711. doi: 10.1146/annurev.immunol.21.120601.141040. [DOI] [PubMed] [Google Scholar]
- 22.Steinman RM, Banchereau J. Taking dendritic cells into medicine. Nature. 2007;449:419–26. doi: 10.1038/nature06175. [DOI] [PubMed] [Google Scholar]
- 23.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. 2004;199:1467–77. doi: 10.1084/jem.20040180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Yamazaki S, Iyoda T, Tarbell K, et al. Direct expansion of functional CD25+CD4+ regulatory T cells by antigen-processing dendritic cells. J Exp Med. 2003;198:235–47. doi: 10.1084/jem.20030422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Tarbell KV, Yamazaki S, Steinman RM. The interactions of dendritic cells with antigen-specific, regulatory T cells that suppress autoimmunity. Semin Immunol. 2006;18:93–102. doi: 10.1016/j.smim.2006.01.009. [DOI] [PubMed] [Google Scholar]
- 26.Liu K, Victora GD, Schwickert TA, et al. In vivo analysis of dendritic cell development and homeostasis. Science. 2009;324:392–7. doi: 10.1126/science.1170540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Waskow C, Liu K, Darrasse-Jeze G, et al. The receptor tyrosine kinase Flt3 is required for dendritic cell development in peripheral lymphoid tissues. Nat Immunol. 2008;9:676–83. doi: 10.1038/ni.1615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Shortman K, Liu YJ. Mouse and human dendritic cell subtypes. Nat Rev Immunol. 2002;2:151–61. doi: 10.1038/nri746. [DOI] [PubMed] [Google Scholar]
- 29.Villadangos JA, Schnorrer P. Intrinsic and cooperative antigen-presenting functions of dendritic-cell subsets in vivo. Nat Rev Immunol. 2007;7:543–55. doi: 10.1038/nri2103. [DOI] [PubMed] [Google Scholar]
- 30.Iyoda T, Shimoyama S, Liu K, et al. The CD8+ dendritic cell subset selectively endocytoses dying cells in culture and in vivo. J Exp Med. 2002;195:1289–302. doi: 10.1084/jem.20020161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.den Haan JM, Lehar SM, Bevan MJ. CD8+ but not CD8- dendritic cells cross-prime cytotoxic T cells in vivo. J Exp Med. 2000;192:1685–96. doi: 10.1084/jem.192.12.1685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Dudziak D, Kamphorst AO, Heidkamp GF, et al. Differential antigen processing by dendritic cell subsets in vivo. Science. 2007;315:107–11. doi: 10.1126/science.1136080. [DOI] [PubMed] [Google Scholar]
- 33.Pooley JL, Heath WR, Shortman K. Cutting edge: intravenous soluble antigen is presented to CD4 T cells by CD8- dendritic cells, but cross-presented to CD8 T cells by CD8+ dendritic cells. J Immunol. 2001;166:5327–30. doi: 10.4049/jimmunol.166.9.5327. [DOI] [PubMed] [Google Scholar]
- 34.Klein L, Hinterberger M, Wirnsberger G, Kyewski B. Antigen presentation in the thymus for positive selection and central tolerance induction. Nat Rev Immunol. 2009;9:833–44. doi: 10.1038/nri2669. [DOI] [PubMed] [Google Scholar]
- 35.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 Med. 2002;196:1627–38. doi: 10.1084/jem.20021598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Schwartz RH. T cell anergy. Annu Rev Immunol. 2003;21:305–34. doi: 10.1146/annurev.immunol.21.120601.141110. [DOI] [PubMed] [Google Scholar]
- 37.Perez VL, Van Parijs L, Biuckians A, Zheng XX, Strom TB, Abbas AK. Induction of peripheral T cell tolerance in vivo requires CTLA-4 engagement. Immunity. 1997;6:411–17. doi: 10.1016/s1074-7613(00)80284-8. [DOI] [PubMed] [Google Scholar]
- 38.Latchman Y, Wood CR, Chernova T, et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2001;2:261–8. doi: 10.1038/85330. [DOI] [PubMed] [Google Scholar]
- 39.Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity. 1999;11:141–51. doi: 10.1016/s1074-7613(00)80089-8. [DOI] [PubMed] [Google Scholar]
- 40.Probst HC, McCoy K, Okazaki T, Honjo T, van den Broek M. Resting dendritic cells induce peripheral CD8+ T cell tolerance through PD-1 and CTLA-4. Nat Immunol. 2005;6:280–6. doi: 10.1038/ni1165. [DOI] [PubMed] [Google Scholar]
- 41.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. Immunity. 2004;20:695–705. doi: 10.1016/j.immuni.2004.05.002. [DOI] [PubMed] [Google Scholar]
- 42.Stranges PB, Watson J, Cooper CJ, et al. Elimination of antigen-presenting cells and autoreactive T cells by Fas contributes to prevention of autoimmunity. Immunity. 2007;26:629–41. doi: 10.1016/j.immuni.2007.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Mellor AL, Munn DH. IDO expression by dendritic cells: tolerance and tryptophan catabolism. Nat Rev Immunol. 2004;4:762–74. doi: 10.1038/nri1457. [DOI] [PubMed] [Google Scholar]
- 44.Munn DH, Mellor AL. Indoleamine 2,3-dioxygenase and tumor-induced tolerance. J Clin Invest. 2007;117:1147–54. doi: 10.1172/JCI31178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Yamazaki S, Dudziak D, Heidkamp GF, et al. CD8+ CD205+ splenic dendritic cells are specialized to induce Foxp3+ regulatory T cells. J Immunol. 2008;181:6923–33. doi: 10.4049/jimmunol.181.10.6923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Coombes JL, Siddiqui KR, Arancibia-Carcamo CV, et al. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-β- and retinoic acid-dependent mechanism. J Exp Med. 2007;204:1757–64. doi: 10.1084/jem.20070590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kretschmer K, Apostolou I, Hawiger D, Khazaie K, Nussenzweig MC, von Boehmer H. Inducing and expanding regulatory T cell populations by foreign antigen. Nat Immunol. 2005;6:1219–27. doi: 10.1038/ni1265. [DOI] [PubMed] [Google Scholar]
- 48.Sun CM, Hall JA, Blank RB, et al. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J Exp Med. 2007;204:1775–85. doi: 10.1084/jem.20070602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Dhodapkar MV, Steinman RM. Antigen-bearing immature dendritic cells induce peptide-specific CD8+ regulatory T cells in vivo in humans. Blood. 2002;100:174–7. doi: 10.1182/blood.v100.1.174. [DOI] [PubMed] [Google Scholar]
- 50.Harnaha J, Machen J, Wright M, et al. Interleukin-7 is a survival factor for CD4+ CD25+ T-cells and is expressed by diabetes-suppressive dendritic cells. Diabetes. 2006;55:158–70. [PubMed] [Google Scholar]
- 51.Hoglund P, Mintern J, Waltzinger C, Heath W, Benoist C, Mathis D. Initiation of autoimmune diabetes by developmentally regulated presentation of islet cell antigens in the pancreatic lymph nodes. J Exp Med. 1999;189:331–9. doi: 10.1084/jem.189.2.331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Gagnerault MC, Luan JJ, Lotton C, Lepault F. Pancreatic lymph nodes are required for priming of β cell reactive T cells in NOD mice. J Exp Med. 2002;196:369–77. doi: 10.1084/jem.20011353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Belz GT, Behrens GM, Smith CM, et al. The CD8α+ dendritic cell is responsible for inducing peripheral self-tolerance to tissue-associated antigens. J Exp Med. 2002;196:1099–104. doi: 10.1084/jem.20020861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Hugues S, Mougneau E, Ferlin W, et al. Tolerance to islet antigens and prevention from diabetes induced by limited apoptosis of pancreatic β cells. Immunity. 2002;16:169–81. doi: 10.1016/s1074-7613(02)00273-x. [DOI] [PubMed] [Google Scholar]
- 55.Kurts C, Cannarile M, Klebba I, Brocker T. Dendritic cells are sufficient to cross-present self-antigens to CD8 T cells in vivo. J Immunol. 2001;166:1439–42. doi: 10.4049/jimmunol.166.3.1439. [DOI] [PubMed] [Google Scholar]
- 56.Chen YG, Choisy-Rossi CM, Holl TM, et al. Activated NKT cells inhibit autoimmune diabetes through tolerogenic recruitment of dendritic cells to pancreatic lymph nodes. J Immunol. 2005;174:1196–204. doi: 10.4049/jimmunol.174.3.1196. [DOI] [PubMed] [Google Scholar]
- 57.Feili-Hariri M, Dong X, Alber SM, Watkins SC, Salter RD, Morel PA. Immunotherapy of NOD mice with bone marrow-derived dendritic cells. Diabetes. 1999;48:2300–8. doi: 10.2337/diabetes.48.12.2300. [DOI] [PubMed] [Google Scholar]
- 58.Feili-Hariri M, Falkner DH, Morel PA. Regulatory Th2 response induced following adoptive transfer of dendritic cells in prediabetic NOD mice. Eur J Immunol. 2002;32:2021–30. doi: 10.1002/1521-4141(200207)32:7<2021::AID-IMMU2021>3.0.CO;2-J. [DOI] [PubMed] [Google Scholar]
- 59.Haase C, Ejrnaes M, Juedes AE, Wolfe T, Markholst H, von Herrath MG. Immunomodulatory dendritic cells require autologous serum to circumvent nonspecific immunosuppressive activity in vivo. Blood. 2005;106:4225–33. doi: 10.1182/blood-2005-03-0975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Creusot RJ, Yaghoubi SS, Kodama K, et al. Tissue-targeted therapy of autoimmune diabetes using dendritic cells transduced to express IL-4 in NOD mice. Clin Immunol. 2008;127:176–87. doi: 10.1016/j.clim.2007.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Perone MJ, Bertera S, Tawadrous ZS, et al. Dendritic cells expressing transgenic galectin-1 delay onset of autoimmune diabetes in mice. J Immunol. 2006;177:5278–89. doi: 10.4049/jimmunol.177.8.5278. [DOI] [PubMed] [Google Scholar]
- 62.Ilarregui JM, Croci DO, Bianco GA, et al. Tolerogenic signals delivered by dendritic cells to T cells through a galectin-1-driven immunoregulatory circuit involving interleukin 27 and interleukin 10. Nat Immunol. 2009;10:981–91. doi: 10.1038/ni.1772. [DOI] [PubMed] [Google Scholar]
- 63.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. 2004;173:4331–41. doi: 10.4049/jimmunol.173.7.4331. [DOI] [PubMed] [Google Scholar]
- 64.Ma L, Qian S, Liang X, et al. Prevention of diabetes in NOD mice by administration of dendritic cells deficient in nuclear transcription factor-κB activity. Diabetes. 2003;52:1976–85. doi: 10.2337/diabetes.52.8.1976. [DOI] [PubMed] [Google Scholar]
- 65.Creusot RJ, Yaghoubi SS, Chang P, et al. Lymphoid-tissue-specific homing of bone-marrow-derived dendritic cells. Blood. 2009;113:6638–47. doi: 10.1182/blood-2009-02-204321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Hackstein H, Thomson AW. Dendritic cells: emerging pharmacological targets of immunosuppressive drugs. Nat Rev Immunol. 2004;4:24–34. doi: 10.1038/nri1256. [DOI] [PubMed] [Google Scholar]
- 67.Moore KW, de Waal Malefyt R, Coffman RL, O'Garra A. Interleukin-10 and the interleukin-10 receptor. Annu Rev Immunol. 2001;19:683–765. doi: 10.1146/annurev.immunol.19.1.683. [DOI] [PubMed] [Google Scholar]
- 68.Tacken PJ, de Vries IJ, Torensma R, Figdor CG. Dendritic-cell immunotherapy: from ex vivo loading to in vivo targeting. Nat Rev Immunol. 2007;7:790–802. doi: 10.1038/nri2173. [DOI] [PubMed] [Google Scholar]
- 69.Mukhopadhaya A, Hanafusa T, Jarchum I, et al. Selective delivery of β cell antigen to dendritic cells in vivo leads to deletion and tolerance of autoreactive CD8+ T cells in NOD mice. Proc Natl Acad Sci USA. 2008;105:6374–9. doi: 10.1073/pnas.0802644105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Bruder D, Westendorf AM, Hansen W, et al. On the edge of autoimmunity: T-cell stimulation by steady-state dendritic cells prevents autoimmune diabetes. Diabetes. 2005;54:3395–401. doi: 10.2337/diabetes.54.12.3395. [DOI] [PubMed] [Google Scholar]
- 71.Jiang W, Swiggard WJ, Heufler C, et al. The receptor DEC-205 expressed by dendritic cells and thymic epithelial cells is involved in antigen processing. Nature. 1995;375:151–5. doi: 10.1038/375151a0. [DOI] [PubMed] [Google Scholar]
- 72.Mahnke K, Guo M, Lee S, et al. The dendritic cell receptor for endocytosis, DEC-205, can recycle and enhance antigen presentation via major histocompatibility complex class II-positive lysosomal compartments. J Cell Biol. 2000;151:673–84. doi: 10.1083/jcb.151.3.673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Kraal G, Breel M, Janse M, Bruin G. Langerhans' cells, veiled cells, and interdigitating cells in the mouse recognized by a monoclonal antibody. J Exp Med. 1986;163:981–97. doi: 10.1084/jem.163.4.981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Swiggard WJ, Mirza A, Nussenzweig MC, Steinman RM. DEC-205, a 205-kDa protein abundant on mouse dendritic cells and thymic epithelium that is detected by the monoclonal antibody NLDC-145: purification, characterization, and N-terminal amino acid sequence. Cell Immunol. 1995;165:302–11. doi: 10.1006/cimm.1995.1218. [DOI] [PubMed] [Google Scholar]
- 75.Inaba K, Swiggard WJ, Inaba M, et al. Tissue distribution of the DEC-205 protein that is detected by the monoclonal antibody NLDC-145. I. Expression on dendritic cells and other subsets of mouse leukocytes. Cell Immunol. 1995;163:148–56. doi: 10.1006/cimm.1995.1109. [DOI] [PubMed] [Google Scholar]
- 76.Witmer-Pack MD, Swiggard WJ, Mirza A, Inaba K, Steinman RM. Tissue distribution of the DEC-205 protein that is detected by the monoclonal antibody NLDC-145. II. Expression in situ in lymphoid and nonlymphoid tissues. Cell Immunol. 1995;163:157–62. doi: 10.1006/cimm.1995.1110. [DOI] [PubMed] [Google Scholar]
- 77.Vremec D, Pooley J, Hochrein H, Wu L, Shortman K. CD4 and CD8 expression by dendritic cell subtypes in mouse thymus and spleen. J Immunol. 2000;164:2978–86. doi: 10.4049/jimmunol.164.6.2978. [DOI] [PubMed] [Google Scholar]
- 78.Henri S, Vremec D, Kamath A, et al. The dendritic cell populations of mouse lymph nodes. J Immunol. 2001;167:741–8. doi: 10.4049/jimmunol.167.2.741. [DOI] [PubMed] [Google Scholar]
- 79.Inaba K, Inaba M, Romani N, et al. Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor. J Exp Med. 1992;176:1693–702. doi: 10.1084/jem.176.6.1693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Granelli-Piperno A, Pritsker A, Pack M, et al. Dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin/CD209 is abundant on macrophages in the normal human lymph node and is not required for dendritic cell stimulation of the mixed leukocyte reaction. J Immunol. 2005;175:4265–73. doi: 10.4049/jimmunol.175.7.4265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Pack M, Trumpfheller C, Thomas D, et al. DEC-205/CD205+ dendritic cells are abundant in the white pulp of the human spleen, including the border region between the red and white pulp. Immunology. 2008;123:438–46. doi: 10.1111/j.1365-2567.2007.02710.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Mahnke K, Qian Y, Knop J, Enk AH. Induction of CD4+/CD25+ regulatory T cells by targeting of antigens to immature dendritic cells. Blood. 2003;101:4862–9. doi: 10.1182/blood-2002-10-3229. [DOI] [PubMed] [Google Scholar]
- 83.DiLorenzo TP, Serreze DV. The good turned ugly: immunopathogenic basis for diabetogenic CD8+ T cells in NOD mice. Immunol Rev. 2005;204:250–63. doi: 10.1111/j.0105-2896.2005.00244.x. [DOI] [PubMed] [Google Scholar]
- 84.Amrani A, Verdaguer J, Serra P, Tafuro S, Tan R, Santamaria P. Progression of autoimmune diabetes driven by avidity maturation of a T-cell population. Nature. 2000;406:739–42. doi: 10.1038/35021081. [DOI] [PubMed] [Google Scholar]
- 85.Han B, Serra P, Amrani A, et al. Prevention of diabetes by manipulation of anti-IGRP autoimmunity: high efficiency of a low-affinity peptide. Nat Med. 2005;11:645–52. doi: 10.1038/nm1250. [DOI] [PubMed] [Google Scholar]
- 86.Bennett CL, Christie J, Ramsdell F, et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet. 2001;27:20–1. doi: 10.1038/83713. [DOI] [PubMed] [Google Scholar]
- 87.Wildin RS, Ramsdell F, Peake J, et al. X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat Genet. 2001;27:18–20. doi: 10.1038/83707. [DOI] [PubMed] [Google Scholar]
- 88.Salomon B, Lenschow DJ, Rhee L, et al. B7/CD28 costimulation is essential for the homeostasis of the CD4+CD25+ immunoregulatory T cells that control autoimmune diabetes. Immunity. 2000;12:431–40. doi: 10.1016/s1074-7613(00)80195-8. [DOI] [PubMed] [Google Scholar]
- 89.Tang Q, Henriksen KJ, Bi M, et al. In vitro-expanded antigen-specific regulatory T cells suppress autoimmune diabetes. J Exp Med. 2004;199:1455–65. doi: 10.1084/jem.20040139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Tarbell KV, Petit L, Zuo X, et al. Dendritic cell-expanded, islet-specific CD4+CD25+CD62L+ regulatory T cells restore normoglycemia in diabetic NOD mice. J Exp Med. 2007;204:191–201. doi: 10.1084/jem.20061631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Arif S, Tree TI, Astill TP, et al. Autoreactive T cell responses show proinflammatory polarization in diabetes but a regulatory phenotype in health. J Clin Invest. 2004;113:451–63. doi: 10.1172/JCI19585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Chentoufi AA, Polychronakos C. Insulin expression levels in the thymus modulate insulin-specific autoreactive T-cell tolerance: the mechanism by which the IDDM2 locus may predispose to diabetes. Diabetes. 2002;51:1383–90. doi: 10.2337/diabetes.51.5.1383. [DOI] [PubMed] [Google Scholar]
- 93.Fife BT, Guleria I, Gubbels Bupp M, et al. Insulin-induced remission in new-onset NOD mice is maintained by the PD-1-PD-L1 pathway. J Exp Med. 2006;203:2737–47. doi: 10.1084/jem.20061577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Kent SC, Chen Y, Bregoli L, et al. Expanded T cells from pancreatic lymph nodes of type 1 diabetic subjects recognize an insulin epitope. Nature. 2005;435:224–8. doi: 10.1038/nature03625. [DOI] [PubMed] [Google Scholar]
- 95.Krishnamurthy B, Dudek NL, McKenzie MD, et al. Responses against islet antigens in NOD mice are prevented by tolerance to proinsulin but not IGRP. J Clin Invest. 2006;116:3258–65. doi: 10.1172/JCI29602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Mannering SI, Harrison LC, Williamson NA, et al. The insulin A-chain epitope recognized by human T cells is posttranslationally modified. J Exp Med. 2005;202:1191–7. doi: 10.1084/jem.20051251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Moriyama H, Abiru N, Paronen J, et al. Evidence for a primary islet autoantigen (preproinsulin 1) for insulitis and diabetes in the nonobese diabetic mouse. Proc Natl Acad Sci USA. 2003;100:10376–81. doi: 10.1073/pnas.1834450100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Nakayama M, Abiru N, Moriyama H, et al. Prime role for an insulin epitope in the development of type 1 diabetes in NOD mice. Nature. 2005;435:220–3. doi: 10.1038/nature03523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Skowera A, Ellis RJ, Varela-Calvino R, et al. CTLs are targeted to kill β cells in patients with type 1 diabetes through recognition of a glucose-regulated preproinsulin epitope. J Clin Invest. 2008;118:3390–402. doi: 10.1172/JCI35449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Steptoe RJ, Ritchie JM, Harrison LC. Transfer of hematopoietic stem cells encoding autoantigen prevents autoimmune diabetes. J Clin Invest. 2003;111:1357–63. doi: 10.1172/JCI15995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Steptoe RJ, Ritchie JM, Jones LK, Harrison LC. Autoimmune diabetes is suppressed by transfer of proinsulin-encoding Gr-1+ myeloid progenitor cells that differentiate in vivo into resting dendritic cells. Diabetes. 2005;54:434–42. doi: 10.2337/diabetes.54.2.434. [DOI] [PubMed] [Google Scholar]
- 102.Toma A, Haddouk S, Briand JP, et al. Recognition of a subregion of human proinsulin by class I-restricted T cells in type 1 diabetic patients. Proc Natl Acad Sci USA. 2005;102:10581–6. doi: 10.1073/pnas.0504230102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Wong FS, Karttunen J, Dumont C, et al. Identification of an MHC class I-restricted autoantigen in type 1 diabetes by screening an organ-specific cDNA library. Nat Med. 1999;5:1026–31. doi: 10.1038/12465. [DOI] [PubMed] [Google Scholar]
- 104.Pociot F, McDermott MF. Genetics of type 1 diabetes mellitus. Genes Immun. 2002;3:235–49. doi: 10.1038/sj.gene.6363875. [DOI] [PubMed] [Google Scholar]
- 105.Vafiadis P, Bennett ST, Todd JA, et al. Insulin expression in human thymus is modulated by INS VNTR alleles at the IDDM2 locus. Nat Genet. 1997;15:289–92. doi: 10.1038/ng0397-289. [DOI] [PubMed] [Google Scholar]
- 106.Tian J, Kaufman DL. Antigen-based therapy for the treatment of type 1 diabetes. Diabetes. 2009;58:1939–46. doi: 10.2337/db09-0451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Kumar P, Ban HS, Kim SS, et al. T cell-specific siRNA delivery suppresses HIV-1 infection in humanized mice. Cell. 2008;134:577–86. doi: 10.1016/j.cell.2008.06.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Peer D, Zhu P, Carman CV, Lieberman J, Shimaoka M. Selective gene silencing in activated leukocytes by targeting siRNAs to the integrin lymphocyte function-associated antigen-1. Proc Natl Acad Sci USA. 2007;104:4095–100. doi: 10.1073/pnas.0608491104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Song E, Zhu P, Lee SK, et al. Antibody mediated in vivo delivery of small interfering RNAs via cell-surface receptors. Nat Biotechnol. 2005;23:709–17. doi: 10.1038/nbt1101. [DOI] [PubMed] [Google Scholar]
- 110.Zheng X, Vladau C, Zhang X, et al. A novel in vivo siRNA delivery system specifically targeting dendritic cells and silencing CD40 genes for immunomodulation. Blood. 2009;113:2646–54. doi: 10.1182/blood-2008-04-151191. [DOI] [PubMed] [Google Scholar]
- 111.Phillips B, Nylander K, Harnaha J, et al. A microsphere-based vaccine prevents and reverses new-onset autoimmune diabetes. Diabetes. 2008;57:1544–55. doi: 10.2337/db07-0507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Atkinson MA, Leiter EH. The NOD mouse model of type 1 diabetes: as good as it gets? Nat Med. 1999;5:601–4. doi: 10.1038/9442. [DOI] [PubMed] [Google Scholar]
- 113.Luo X, Tarbell KV, Yang H, et al. Dendritic cells with TGF-β1 differentiate naive CD4+CD25- T cells into islet-protective Foxp3+ regulatory T cells. Proc Natl Acad Sci USA. 2007;104:2821–6. doi: 10.1073/pnas.0611646104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Oluwole OO, Depaz HA, Gopinathan R, et al. Indirect allorecognition in acquired thymic tolerance: induction of donor-specific permanent acceptance of rat islets by adoptive transfer of allopeptide-pulsed host myeloid and thymic dendritic cells. Diabetes. 2001;50:1546–52. doi: 10.2337/diabetes.50.7.1546. [DOI] [PubMed] [Google Scholar]
- 115.Rastellini C, Lu L, Ricordi C, Starzl TE, Rao AS, Thomson AW. Granulocyte/macrophage colony-stimulating factor-stimulated hepatic dendritic cell progenitors prolong pancreatic islet allograft survival. Transplantation. 1995;60:1366–70. [PMC free article] [PubMed] [Google Scholar]
- 116.Dhodapkar MV, Steinman RM, Krasovsky J, Munz C, Bhardwaj N. Antigen-specific inhibition of effector T cell function in humans after injection of immature dendritic cells. J Exp Med. 2001;193:233–8. doi: 10.1084/jem.193.2.233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Lu W, Arraes LC, Ferreira WT, Andrieu JM. Therapeutic dendritic-cell vaccine for chronic HIV-1 infection. Nat Med. 2004;10:1359–65. doi: 10.1038/nm1147. [DOI] [PubMed] [Google Scholar]
- 118.Giannoukakis N, Phillips B, Trucco M. Toward a cure for type 1 diabetes mellitus: diabetes-suppressive dendritic cells and beyond. Pediatr Diabetes. 2008;9:4–13. doi: 10.1111/j.1399-5448.2008.00401.x. [DOI] [PubMed] [Google Scholar]
- 119.Thomson AW, Robbins PD. Tolerogenic dendritic cells for autoimmune disease and transplantation. Ann Rheum Dis. 2008;67(Suppl 3):iii90–6. doi: 10.1136/ard.2008.099176. [DOI] [PubMed] [Google Scholar]
- 120.Hamilton-Williams EE, Martinez X, Clark J, et al. Expression of diabetes-associated genes by dendritic cells and CD4 T cells drives the loss of tolerance in nonobese diabetic mice. J Immunol. 2009;183:1533–41. doi: 10.4049/jimmunol.0900428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Steptoe RJ, Ritchie JM, Harrison LC. Increased generation of dendritic cells from myeloid progenitors in autoimmune-prone nonobese diabetic mice. J Immunol. 2002;168:5032–41. doi: 10.4049/jimmunol.168.10.5032. [DOI] [PubMed] [Google Scholar]
- 122.Vasquez AC, Feili-Hariri M, Tan RJ, Morel PA. Qualitative and quantitative abnormalities in splenic dendritic cell populations in NOD mice. Clin Exp Immunol. 2004;135:209–18. doi: 10.1111/j.1365-2249.2003.02359.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Weaver DJ, Jr, Poligone B, Bui T, Abdel-Motal UM, Baldwin AS, Jr, Tisch R. Dendritic cells from nonobese diabetic mice exhibit a defect in NF-kappa B regulation due to a hyperactive I kappa B kinase. J Immunol. 2001;167:1461–8. doi: 10.4049/jimmunol.167.3.1461. [DOI] [PubMed] [Google Scholar]
- 124.Shultz LD, Ishikawa F, Greiner DL. Humanized mice in translational biomedical research. Nat Rev Immunol. 2007;7:118–30. doi: 10.1038/nri2017. [DOI] [PubMed] [Google Scholar]
