Dendritic cell immunotherapy for autoimmune diabetes | SpringerLink

16 downloads 0 Views 132KB Size Report
Dendritic cells (DC) play important roles in the initiation of immune responses and maintenance of self-tolerance. We have been studying the role of DC in the ...
Immunologic Research 2006;36/1–3:167–173

Dendritic Cell Immunotherapy for Autoimmune Diabetes

Abstract Dendritic cells (DC) play important roles in the initiation of immune responses and maintenance of self-tolerance. We have been studying the role of DC in the pathogenesis of type 1 diabetes and exploring the ability of specific DC subsets to prevent diabetes in non-obese diabetic (NOD) mice. DC subsets that prevent diabetes in this model have a mature phenotype and induce the production of regulatory Th2 cells. We review here recent advances in this area and highlight the importance of optimizing culture conditions and purification methods in the isolation of therapeutic DC.

Maryam Feili-Hariri1,3 Rafael R. Flores3 A. Cecilia Vasquez3 Penelope A. Morel2,3

Key Words Dendritic cells Type 1 diabetes Autoimmunity Immunotherapy

Introduction Over the last several years the work in our laboratory has focused on the study of dendritic cells in the murine model of type 1 diabetes (T1D), the non-obese diabetic (NOD) mouse. T1D is an autoimmune disease char-

Departments of 1Surgery, 2 Medicine, and 3Immunology, University of Pittsburgh, Pittsburgh, PA

acterized by the destruction of the insulinproducing β cells of the islets of Langerhans (1). Prior to the development of diabetes, the islets become heavily infiltrated with lymphoid cells, including CD4+, CD8+ T cells, dendritic cells, and monocytes (2,3) and by the time diabetes appears over 90% of the

Penelope A. Morel, MD Department of Immunology, University of Pittsburgh, 200 Lothrop Street, BST E1048, Pittsburgh, PA 15261. E-mail: [email protected] and Maryam Feili-Hariri, PhD Departments of Surgery and Immunology, University of Pittsburgh, 3550 Terrace Street, 666 Scaife Hall, Pittsburgh, PA 15261. E-mail: [email protected]

© 2006 Humana Press Inc. 0257–277X/ (Online)1559-0755/06/ 36/1–3:167–173/$30.00

167

islets have been destroyed. Therapeutic interventions to prevent diabetes have to be aimed at the period of insulitis during which most of the destruction takes place. The NOD mouse is a good model of human T1D as it shares many of the genetic and immunological features of the human disease (4). NOD mice (females>males) spontaneously develop diabetes between 15 and 20 wk of age (4). Genetic analysis of diabetes susceptibility in the NOD mouse has revealed that a minimum of 15 genes are implicated, and one of these is mapped to the MHC (5). Many of the other genes associated with T1D have important functions in the immune system, and include genes important in T cell differentiation and function (5). It has been known for some time that effector Th1 cells and CTL are responsible for the autoimmunity in NOD mice (6–8). This effect seems to be both at the level of the initiation of the autoimmune response and at the stage of islet cell destruction. The importance of the Th1 pathway has been demonstrated in various knockout systems in which it was shown that IRF1–/– (9) and Stat4–/– NOD mice (10) were protected from the development of diabetes. In contrast Stat 6–/– NOD mice developed diabetes more rapidly than wild-type NOD suggesting that endogenous Th2 cells can protect mice to some degree (10). We have been interested in determining whether specific DC subsets play a role in the development of the type 1 skewed immune response in NOD mice and we have explored the use of specific DC subsets in the prevention of diabetes onset. Do DC Defects Contribute to the Development of Autoimmunity in NOD Mice? In T1D, several groups have reported abnormalities in DC phenotype and function

168

in both the mouse and human, which may result in the skewing of the response toward pathogenic Th1 cells (11–15). Studies on bone marrow (BM)–derived DC have been somewhat controversial with some groups reporting increases in IL-12p70 production (11–13,15), a cytokine important for Th1 differentiation, while others including ourselves have reported reduced IL-12p70 production by NOD BMderived DC (16–18). These discrepancies led us to the detailed examination of the phenotype and function of BM DC in NOD mice. There is agreement among all investigators that there is a defect in the number of DC generated from NOD BM (13,14,16,18), which appears to be related to a defect in the responsiveness of NOD BM cells to the cytokine GM-CSF (19). However, there is disagreement as to whether NOD DC are more or less prone to drive Th1 differentiation, or T cell activation. Some of these discrepancies could be due to differences in the culture conditions used in the generation of BM-derived DC, which vary in terms of cytokine concentrations used, length of the culture, and method of purification. We have found that all of these factors can have a profound influence on DC phenotype and function. We have conducted experiments in which we have tested different culture systems and maturation conditions. In these experiments we compared DC from NOD and BALB/c mice generated either in the presence of GM-CSF (GM DC) alone or GMCSF + IL-4 (GM4 DC). The GM DC were induced to mature with either CpG or TNF-α + PGE2. Following maturation the DC were stimulated with J-558 cells expressing CD40L in the presence of IL-4 and the resulting supernatants were analyzed for the presence of IL12p70 (Fig. 1). It was apparent that some conditions, such as CpG, led to increased IL12p70 production by NOD DC, whereas other conditions, such as GM4 DC, resulted in increased IL-12p70 production by BALB/c

Feili-Hariri et al.

Fig. 1. Maturation cocktail influences ability of DC to produce IL-12p70. BM DCs were generated in the presence of GM-CSF for 4 d. On d 4 GM DCs were matured with TNF-α + PGE2 or CpG 1668 (PS) overnight. Simultaneously, BM DCs were also generated in the presence of GM-CSF and IL-4 for 5 d. On d 5 all DCs were harvested using an anti-CD11c Mini-MACS column. GM DCs were transferred into 48 well plates at a concentration of 1× 106 cells/mL and co-cultured for 24 h with CD40L expression J-558 cells at a concentration of 1× 106 cells/mL plus IL-4 (2 ng/mL). The supernatants were analyzed for the presence of IL-12p70 by ELISA. The data presented are representative of two independent experiments.

DC (Fig. 1). These data suggest that the study of BM-derived DC in NOD mice may not be representative of what occurs in vivo, and furthermore the plasticity of DC progenitors from NOD mice can lead to the generation of DCs with either Th1 or Th2 promoting activity (20). Thus, our laboratory and other investigators have performed studies to examine the phenotype and function of DC subsets in vivo. It has been observed that NOD mice exhibit defects in the number of regulatory T cells, including NKT cells, CD4+ CD25+ T cells, and Th2 cells (21–23). Recent studies have implicated several DC subsets in the maintenance and activation of different Treg populations (24). Several groups including our own have determined that NOD mice have an imbalance in the numbers of specific splenic DC subsets. This is characterized by a relative decrease in the number of CD8α+ DC (14,18) and an increase in the number of myeloid DC. We have also observed that splenic DC from NOD mice produce lower levels of IL-12p40 than similar populations from C57BL/6 mice (18). In a study from O’Keeffe et al., the defect in

Dendritic Cells and Autoimmunity

CD8α+ DC numbers was mapped to a nonMHC susceptibility locus on chromosome 4 (25), and these investigators also observed a reduction in the levels of IL-12 produced by NOD DC. Recent studies targeting antigens to CD8α+ DC through the use of the antiDEC205 mAb have resulted in either the deletion of antigen-specific T cells (26) or the induction of specific Tregs (27), demonstrating a role for these DCs in the maintenance of steady-state tolerance. We observed that the administration of recombinant human Flt3 ligand (FL) to NOD mice preferentially increased the number of CD8α+ DC (18). We also observed that both CD8α+ and CD8α– DCs could delay the onset of diabetes and induce some degree of protection from diabetes development (18). O’Keeffe et al. followed this up by demonstrating that FL treatment could prevent diabetes in NOD mice (25). These data collectively suggest that NOD mice have a defect in the generation of specific DC subsets that may play an important role in the development and maintenance of self-tolerance.

169

DCs Can Be Used to Correct the Autoimmune Response In parallel we have also examined the ability of BM-derived DC to prevent and/or treat diabetes. When we started these studies several years ago, we anticipated that so-called immature DC (iDC), expressing low levels of costimulatory molecules and MHC class II, would be more effective in diabetes prevention than mature/stimulatory DC (mDC). To our surprise, we observed that mDC were able to prevent disease when given to young prediabetic NOD mice, whereas iDC were completely ineffective (28). The therapeutic DCs expressed high levels of CD80, CD86, and CD40 and, when stimulated using CD40L or LPS, produced low levels of IL-12p70 (16). Of note, the most mature DCs were the best at preventing the disease. In a study examining the role of gene-modified DCs using adenoviral vectors, we observed that a single injection of adenovirally infected DCs given at 8 wk of age could protect all NOD mice from developing diabetes (29). DCs infected with adenoviral vectors expressed a high level of maturation markers. Although a few investigators have reported modest success with iDC (30), most reports of successful DC therapy in NOD mice have involved the use of DCs matured using various culture conditions (31–33). This may be explained by the fact that the development and maintenance of Tregs require high levels of co-stimulation and the presence of IL-2. We analyzed the mechanism by which mDC prevent diabetes in this model and our early observation suggested that the DC treatment induced regulatory Th2 cells (34). Indeed, splenocytes from NOD mice that were given DCs 7 wk prior to in vitro activation produced high levels of the type 2 cytokines IL-4 and IL-5, whereas splenocytes from age-matched untreated NOD mice produced undetectable levels of these cytokines

170

Fig. 2. Analysis of CD45RBlo CD25+ CD4+ T cells in the spleen of (A) 12-wk-old mice treated with DCpep (DC, n=10) or PBS (n=5), or (B) 32-wk-old nondiabetic mice treated with DCpep (n=4) and mice treated with DCmed (n=4). Seven-week-old prediabetic (Pre, n=3) and 24–28-wk-old diabetic (Diab, n=5) mice were included as controls. Cells were stained with rat anti-mouse FITC-conjugated CD45RB, PE-conjugated CD25, and Cychrome-conjugated CD4 mAbs or isotype controls for 30 min on ice. Small CD4+ T cells were analyzed for the expression of CD45RBlo/CD25+ cells by flow cytometry.

(34). In addition, we observed an increase in the number of CD4+ CD25+, CD45RBlo T cells in the spleen of DC-treated mice (Fig. 2). These cells were sorted and found to secrete type 2 cytokines following activation, whereas they failed to suppress the proliferation of CD4+ CD25– T cells in a classic suppression assay (34). At the time that we performed these studies, the FoxP3 mAb was not available. It is possible that Tregs were also induced in addition to the Th2 effectors and/or expanded by the DC treatment but we were not able to detect them. We are actively pursuing this possibility at the present time. This speculation is supported by the recent demonstration by Tarbell et al. that sorted CD86hi DC from NOD mice can be used to expand sorted CD25+ Treg in vitro and that these expanded Tregs had suppressor activity in

Feili-Hariri et al.

Fig. 3. The influence of the method of purification on DC phenotype and function. DCs were generated from BM progenitors in the presence of GM-CSF+ IL-4. The GM+ IL-4 DCs were purified on 14.5% metrizamide gradients (solid lines) or DC were purified using CD11c beads (dashed lines). (A) The Purified DCs were stained with the indicated mAbs and were analyzed by flow cytometry on an open gate. (B) Purified DCs were activated with LPS + IFN-γ for 24 h and culture supernatant were collected and analyzed for IL-12p70 by ELISA.

vitro and prevented diabetes development in prediabetic NOD mice (35). The fact that sorted CD86hi DC could be used to expand preexisting Tregs was interesting and prompted us to further examine the therapeutic DCs in our system. The DCs that we used were isolated following a relatively short (4–5 d) culture of BM progenitors in the presence of GM-CSF and IL-4 (28). At the end of the culture the cells were purified on a 14.5% metrizamide gradient and the cells routinely expressed a uniformly high level of CD86 and other costimulatory molecules (16,28,29). More recently, we have compared the use of CD11c microbeads with the metrizamide gradient, and we have observed a significant difference in the phenotype and function of DCs isolated using these two techniques (Fig. 3). When GM4 DCs were purified using CD11c beads, two populations were clearly distinguishable, a minor population that expressed high levels of CD86 representing mDC and a

Dendritic Cells and Autoimmunity

larger population of iDC. The metrizamide purification specifically enriched for the CD86hi DCs (Fig. 3). Interestingly, the metrizamidepurified DC produced lower levels of IL-12p70 compared to the DCs isolated using CD11c microbeads. This difference was not due to the ability of metrizamide to induce DC maturation because in our early studies we used a 16% metrizamide gradient to purify the iDC, and these cells maintained a very immature phenotype following the purification step (28). We believe that the use of metrizamide gradients allowed us to “sort” CD86hi DC from the BM cultures, thus isolating a DC population with an increased ability to stimulate type 2 responses, but also perhaps Tregs. Our more recent studies have focused on the identification of other phenotypic markers or secreted cytokines/chemokines that may be playing a role in the therapeutic effect of mDC in NOD mice. Microarray analysis comparing the gene expression profiles of the

171

mDC and iDC used in these studies have revealed several additional molecules that are expressed more highly in the therapeutic DC compared to iDC. These include OX40L, CD200, and various chemokine and secreted proteins thought to be associated with the development of Th2 cells or Tregs. Future Prospects for DC Therapy Several issues need to be addressed before DC therapy can be considered for use in human prediabetics. The DC therapies that we have used are most effective when given early during the prediabetic phase, and clearly strategies should be developed such that this could be given later once symptoms have started. We have shown that DCs transduced to express the cytokine IL-4 can be given much later, once destructive insulitis has begun (29). We are presently exploring using DCs that produce IL-10 or TGF-β in addition to IL-4 with the hopes of inducing additional Treg populations, and thereby allowing us to delay treatment even longer. Another is the issue of antigen specificity. In our system we have found that unpulsed DCs are as effective in preventing disease as those pulsed with islet-derived antigens or peptides (28). We have explained this by demonstrating that DCs migrate to both pancreas and pancreatic lymph

node (28,29) and speculating that the DCs acquire specific antigen at these sites, although this remains to be proven. However, a recent publication has demonstrated that this could be related to the use of fetal calf serum (FCS) in the culture medium (36). These investigators demonstrated that DCs grown in FCS induced a systemic immune deviation, whereas the culture of DC in autologous mouse serum could induce antigen-specific immunomodulation (36). The DCs grown in mouse serum were only modestly effective in preventing disease, and this may be due to the low levels of CD86 expressed by these cells. In our own experience the culture of BM cells in the presence of mouse serum does not give rise to DCs with the same degree of maturity as those grown in FBS. In our view it is important to identify those characteristic features of DC that are important for the therapeutic effect and then focus on developing culture conditions in the human to optimize the generation of DC with the ability to stimulate antigen-specific Tregs of various types. Acknowledgments The authors would like to thank Dewayne Falkner for expert technical assistance. This work was supported by NIH grant CA73743 (PAM) and DOD training grant DAMD17-991-9352 (RRF).

References 1. Castano L, Eisenbarth GS: Type-I diabetes: a chronic autoimmune disease of human, mouse and rat. Annu Rev Immunol 1990;8:647–679. 2. Jansen A, Homo-Delarche F, Hooijkaas H, Leenen PJ, Dardenne M, Drexhage HA: Immunohistochemical characterization of monocytes-macrophages and dendritic cells involved in the initiation of the insulitis and β-cell destruction in NOD mice. Diabetes 1994;43:667–675. 3. Miyazaki A, Hanafusa T, Yamada J, et al: Predominance of T lymphocytes in pancreatic islets and spleen of pre-

172

diabetic nonobese diabetic (NOD) mice: a longitudinal study. Clin Exp Immunol 1985;6:622–625. 4. Leiter EL, Prochazka M, Coleman DL: Animal model of human disease, the nonobese diabetic (NOD) mouse. Am J Pathol 1987;128:380. 5. Todd JA, Wicker LS: Genetic protection from the inflammatory disease type 1 diabetes in humans and animal models. Immunity 2001;15:387–395. 6. Christianson SW, Schultz LD, Leiter ED: Adoptive transfer of diabetes into immunodeficient NOD-scid/scid

Feili-Hariri et al.

7.

8. 9.

10. 11.

12. 13.

14. 15.

16. 17.

18.

19. 20.

21. 22.

mice. Relative contributions of CD4+ and CD8+ T cells from diabetic versus prediabetic NOD.NON-Thy-1a donors. Diabetes 1993;42:44–55. Soderstrom I, Bergman ML, Colussi F, Lejon K, Bergqvist I, Holmberg D: Establishment and characterization of RAG-2 deficient non-obese diabetic mice. Scand J Immunol 1996;43:525–530. Wang B, Gonzalez A, Benoist C, Mathis D: The role of CD8+ T cells in the initiation of insulin-dependent diabetes mellitus. Eur J Immunol 1996;26:1762–1769. Nakazawa T, Satoh J, Takahashi K, et al: Complete suppression of insulitis and diabetes in NOD mice lacking interferon regulatory factor-1. J Autoimmun 2001;17: 119–125. Chatenoud L, Salomon B, Bluestone JA: Suppressor T cells—they’re back and critical for regulation of autoimmunity. Immunol Rev 2001;182:149–163. Liu J, Beller D: Aberrant production of IL-12 by macrophages from several autoimmune-prone mouse strains is characterized by intrinsic and unique patterns of NF-kappa B expression and binding to the IL-12 p40 promoter. J Immunol 2002;169:581–586. Marleau AM, Singh B: Myeloid dendritic cells in nonobese diabetic mice have elevated costimulatory and T helper-1-inducing abilities. J Autoimmun 2002;19:23–35. Poligone B, Weaver DJJ, Sen P, Baldwin ASJ, Tisch R: Elevated NF-kappaB activation in nonobese diabetic mouse dendritic cells results in enhanced APC function. J Immunol 2002;168:188–196. Prasad SJ, Goodnow CC: Cell-intrinsic effects of nonMHC NOD genes on dendritic cell generation in vivo. Int Immunol 2002;14:677–684. Steptoe RJ, Ritchie JM, Harrison LC: Increased generation of dendritic cells from myeloid progenitors in autoimmune-prone nonobese diabetic mice. J Immunol 2002;168:5032–5041. Feili-Hariri M, Morel PA: Phenotypic and functional characteristics of BM-derived DC from NOD and non diabetes-prone strains. Clin Immunol 2001;98:133–142. Strid J, Lopes L, Marcinkiewicz J, et al: A defect in bone marrow derived dendritic cell maturation in the nonobesediabetic mouse. Clin Exp Immunol 2001;123: 375–381. 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;135:209–218. Langmuir PB, Bridgett MM, Bothwell AL, Crispe IN: Bone marrow abnormalities in the non-obese diabetic mouse. Int Immunol 1993;5:169–177. Feili-Hariri M, Falkner DH, Morel PA: Polarization of naive T cells into Th1 or Th2 by distinct cytokine-driven murine dendritic cell populations: implications for immunotherapy. J Leukoc Biol 2005;78:656–664. Baxter AG, Kinder SJ, Hammond KJ, Scollay R, Godfrey DI: Association between alphabetaTCR+CD4–CD8– T-cell deficiency and IDDM in NOD/Lt mice. Diabetes 1997;46:572–582. Poulton LD, Smyth MJ, Hawke CG, et al: Cytometric and functional analyses of NK and NKT cell deficiencies in NOD mice. Int Immunol 2001;13:887–896.

Dendritic Cells and Autoimmunity

23. 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;12:431–440. 24. Groux H, Fournier N, Cottrez F: Role of dendritic cells in the generation of regulatory T cells. Sem Immunol 2004;16:99–106. 25. O’Keeffe M, Brodnicki TC, Fancke B, et al: Fms-like tyrosine kinase 3 ligand administration overcomes a genetically determined dendritic cell deficiency in NOD mice and protects against diabetes development. Int Immunol 2005;17:307–314. 26. 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: 769–780. 27. 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;6:1219–1227. 28. 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; 48:2300–2308. 29. Feili-Hariri M, Falkner DH, Gambotto A, et al: Dendritic cells transduced to express IL-4 prevent diabetes in nonobese diabetic mice with established insulitis. Human Gene Ther 2003;14:13–23. 30. 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;173:4331–4341. 31. Naumov YN, Bahjat KS, Gausling R, et al: Activation of CD1d-restricted T cells protects NOD mice from developing diabetes by regulating dendritic cell subsets. Proc Natl Acad Sci USA 2001;98:13838–13843. 32. Papaccio G, Nicoletti F, Pisanti FA, Bendtzen K, Galdieri M: Prevention of spontaneous autoimmune diabetes in NOD mice by transferring in vitro antigenpulsed syngeneic dendritic cells. Endocrinology 2000; 141:1500–1505. 33. Shinomiya M, Fazle Akbar SM, Shinomiya H, Onji M: Transfer of dendritic cells (DC) ex vivo stimulated with interferon-gamma (IFN-gamma) down-modulates autoimmune diabetes in non-obese diabetic (NOD) mice. Clin Exp Immunol 1999;117:38–43. 34. 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;32:2021–2030. 35. 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;199:1467–1477. 36. 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: 4225–4233.

173