Abstract
The non-obese diabetic (NOD) mouse model of autoimmune (type 1) diabetes has contributed greatly to our understanding of disease pathogenesis and has facilitated the development and testing of therapeutic strategies to combat the disease. Although the model is a valuable immunological tool in its own right, it reaches its fullest potential in areas where its findings translate to the human disease. Perhaps the foremost example of this is the field of T-cell antigen discovery, from which diverse benefits can be derived, including the development of antigen-specific disease interventions. The majority of NOD T-cell antigens are also targets of T-cell autoimmunity in patients with type 1 diabetes, and several of these are currently being evaluated in clinical trials. Here we review the journeys of these antigens from bench to bedside. We also discuss several recently identified NOD T-cell autoantigens whose translational potential warrants further investigation.
Keywords: autoantigens, autoimmune diabetes, non-obese diabetic mice, therapies, type 1 diabetes
Introduction
The non-obese diabetic (NOD) mouse is a widely studied model for spontaneous autoimmune (type 1) diabetes that is polygenic and T-cell-mediated.1 However, those new to the field are often unaware of its serendipitous origin.2,3 In the 1970s, outbred Imperial Cancer Research mice were being selectively bred to derive a strain characterized by cataracts [which later became the cataract Shionogi (CTS) strain]. At the sixth generation, two sublines were separately maintained, one exhibiting fasting hyperglycaemia and the other euglycaemia. Ironically, a single female in the latter line developed diabetes at the 20th generation. Her progeny, born during her prediabetic period, were intercrossed, and mice having one or two parents that developed diabetes were selected for subsequent rounds of breeding. This selective breeding for diabetes eventually led to the establishment of the now inbred NOD strain. The genetic loci that contribute to disease in these mice have recently been catalogued and discussed.4 Here we will focus our attention on the diabetes-related autoantigens that are targeted by T cells in the NOD strain. For at least 20 years, identification of these antigens has been the goal of numerous laboratories, including our own. These efforts have yielded 18 antigens to date (Table 1),5 supporting the idea that multiple antigens contribute to the pathogenesis of type 1 diabetes. Here we will discuss primarily those that have been used in antigen-specific type 1 diabetes prevention or intervention trials in humans. Several recently identified NOD T-cell antigens whose potential for translation has not yet been fully explored will also be highlighted.
Table 1.
Diabetes-relevant T-cell antigens in non-obese diabetic (NOD) mice
| Antigen | Reference |
|---|---|
| Heat-shock protein 60 | 6 |
| Glutamic acid decarboxylase 65 | 26 |
| Carboxypeptidase E | 26 |
| Peripherin | 26 |
| Insulin | 67 |
| Glutamic acid decarboxylase 67 | 87 |
| Islet cell autoantigen 69 | 88 |
| Insulinoma-associated protein 2β | 89 |
| Glucose transporter 2 | 90 |
| Insulinoma-associated protein 2 | 91 |
| Hepatocarcinoma-intestine-pancreas/pancreatic associated protein | 70 |
| S100β | 92 |
| Glial fibrillary acidic protein | 92 |
| Islet-specific glucose-6-phosphatase catalytic subunit-related protein | 93 |
| Dystrophia myotonica kinase | 94 |
| Regenerating gene II | 68 |
| Pancreatic duodenal homeobox 1 | 72 |
| Chromogranin A | 73 |
Antigens are listed in chronological order of their description as T-cell targets in NOD mice.
NOD T-cell antigens: from bench to bedside
Heat-shock protein 60 (Hsp60)
Two decades ago,6 Cohen and colleagues reported the presence in NOD mice of serum antibodies to the 65-kD heat-shock protein of Mycobacterium tuberculosis (Mtb Hsp65). Proliferation of splenic T cells from NOD mice in response to Mtb Hsp65 was also observed, and CD4 T-cell clones specific for the protein could induce diabetes when transferred to young (4-week-old) female NOD mice.6 The investigators speculated that these antibody and T-cell reactivities reflected an autoimmune response to a cellular protein related to Mtb Hsp65. Subsequent work7 revealed that the diabetogenic Mtb Hsp65-specific T-cell clones recognized a peptide derived from human Hsp60. The peptide corresponds to amino acids 437–460 of human Hsp60 and is referred to throughout the literature as p277. Murine and human p277 differ at only a single position (Fig. 1), and NOD-derived T cells respond equivalently to the two peptides.8 Remarkably, a single administration of human p277 in incomplete Freund's adjuvant to 4-week-old NOD mice reduced the incidence of spontaneous diabetes at 32 weeks of age from 80% to 31%.7 Significant benefit was observed even when the mice were treated later in the disease process (i.e., at 12, 15 or 17 weeks of age).9
Figure 1.

Sequence comparison of human and mouse heat-shock protein 60 (Hsp60) p277, DiaPep277, and the related peptide from Mycobacterium tuberculosis Hsp65. Dashes indicate identity to human p277, which corresponds to amino acids 437–460 of Hsp60. DiaPep277 is the version of human Hsp60 p277 that is used in clinical trials.
These encouraging preclinical results, coupled with the finding that type 1 diabetes patients show T-cell proliferative responses to Hsp60 peptides including p277,9,10 inspired a series of still-ongoing clinical trials to explore the efficacy of p277 in the treatment of type 1 diabetes in humans. In the clinical trials, a modified version of the peptide (DiaPep277) is employed in which the two cysteines are substituted with valine for improved stability (Fig. 1).11 The immunological activity of DiaPep277 is identical to that of the native peptide.12 Results of a phase II trial of DiaPep277 were first reported in 2001.11 Trial participants were newly diagnosed (within 6 months) with type 1 diabetes and ranged in age from 16 to 58 years. Patients were treated at study entry, and at 1, 6 and 12 months, with 1 mg DiaPep277. While mean glucagon-stimulated C-peptide concentrations dropped in the placebo group, they were maintained in the DiaPep277 group at both 10 months11 and 18 months13 after study entry, suggesting preservation of beta cell function. Similar findings were obtained by others in patients receiving 2·5 mg of the peptide.14 While studies in children with type 1 diabetes have been less encouraging,15,16 benefit in some children has been suggested.16 The potential of DiaPep277 to preserve beta cell function in type 1 diabetes patients is currently being further explored in two ongoing phase III trials (NCT00615264 and NCT01103284).17
The mechanism of action of DiaPep277 appears to be multifaceted. The peptide can bind the NOD class II major histocompatibility complex (MHC) molecule I-Ag7,18 and presumably also human leucocyte antigen (HLA)-DQ8, as these two MHC molecules bind a similar spectrum of peptides.19 Subcutaneous administration of DiaPep277 to NOD mice results in transient activation of peptide-specific T cells that produce the T helper type 2 (Th2) cytokines interleukin (IL)-4 and IL-10.20 However, additional mechanisms of action for DiaPep277 have also been suggested.21,22 Treatment of purified mouse or human T cells with the peptide results in inhibition of T-cell chemotaxis, and this effect is dependent on Toll-like receptor 2 (TLR2) signalling.21 The peptide also enhances the suppressive activity of human regulatory T cells in a TLR2-dependent manner.22
Glutamic acid decarboxylase (GAD)
GAD was one of the earliest identified autoantigens in type 1 diabetes.23 GAD is an enzyme involved in the production of the neurotransmitter γ-aminobutyric acid (GABA), and is expressed exclusively in the brain and pancreas. It exists as one of two distinctly encoded isoforms, GAD65 and GAD67, with GAD65 reactivity primarily associated with type 1 diabetes. The physiological role of GAD in the pancreas is unclear, although GABA is thought to be involved in the regulation of insulin secretion.24
GAD was initially identified as an autoantigen using serum antibodies from patients with type 1 diabetes.23 It was later identified as a major early target of islet-reactive T cells in NOD mice.25,26 Furthermore, studies have demonstrated that GAD65-reactive cells can directly damage the pancreatic islets.27 The early involvement of GAD65 in the disease course suggests that GAD may play an important and possibly fundamental role in the disease pathogenesis. The importance of GAD65 in type 1 diabetes has been further reinforced by a large set of in vivo studies performed in NOD mice. As the inflammatory process begins in earnest at about 3–4 weeks of age in NOD mice, immunotherapy of young mice with GAD65 protein or peptides was thought to be a promising approach. Indeed, numerous studies have demonstrated the efficacy of GAD65 treatment in preventing both insulitis and diabetes.25,26,28–30 The beneficial effect of treatment with GAD65 is not limited to young mice, but extends even to older mice in which the inflammatory process is well underway.31,32 Additionally, novel strategies for delivering GAD have been developed, including plasmid DNA vaccination,33 recombinant vaccinia virus administration,34 and a gene gun-mediated DNA vaccination approach.35
Understanding the role of GAD65 is a critical question in type 1 diabetes and it has been theorized that the Th1 subset of CD4 T cells is responsible for much of the beta cell damage, while the Th2 subset is associated with immunoprotective qualities. Many of the treatment studies with GAD65 have demonstrated that the CD4 T-cell profile shifts from Th1 to Th2 following treatment,26,30,31,36 and the protection from insulitis and diabetes is thought to be a consequence of this shift, at least in part. Additionally, GAD65 treatment appears to induce regulatory T-cell populations that can suppress disease.30–32,36–38 It appears that, while certain populations of the GAD65-reactive CD4 T cells are indeed diabetogenic, others play an important immunoregulatory role.
While there appears to be no question that GAD65-reactive T cells play an important role in the disease process, it remains unclear if GAD is an essential antigen. The oft-replicated prevention of type 1 diabetes through the treatment of GAD65 suggests that reactivity to GAD65 plays a critical role in the disease pathogenesis. Furthermore, a transgenic NOD mouse model lacking GAD65 expression in the islets was prevented from developing insulitis and diabetes.39 However, further studies have indicated that the disease process proceeds normally in NOD mice even in the absence of GAD65 expression.40 Also, widespread expression of GAD65 and the presumed development of tolerance did not affect insulitis or diabetes development.41 When taken together, these studies suggest a need for further clarification of the contribution of GAD65 to the pathogenesis of type 1 diabetes.
GAD65 (and not GAD67) is expressed in human islets, and many of the observations in NOD mice are proposed to apply to type 1 diabetes in humans as well. As the initial detection of GAD65 as an antigen occurred in humans, translating the treatment successes from NOD mice back to humans would allow the research arc to come full circle. Diamyd, recombinant human GAD65 formulated with alum, has successfully progressed though phase II trials in patients having latent autoimmune diabetes in adults (LADA)42,43 or recent-onset type 1 diabetes44 and has shown promise in preserving beta cell function. When type 1 diabetes patients, ranging in age from 10 to 18 years, were treated with Diamyd within 6 months of diagnosis (two subcutaneous injections, 1 month apart), they showed less of a decline in both fasting and stimulated C-peptide levels after 30 months than did the placebo-treated group.44 Diamyd is currently in large-scale phase III trials in newly diagnosed type 1 diabetes patients in the USA (NCT00751842) and Europe (NCT00723411).
Preproinsulin
Insulin is a major autoantigen in both NOD mice and human patients.45 There are two insulin genes expressed in mice, Ins1 and Ins2. T-cell responses are generated against peptides from both the mature insulin and the cleaved signal and C peptides.46 Although both genes are expressed in the islets, only Ins2 appears to be expressed in the thymus.47 This differential expression is believed to be responsible for the findings that Ins1 knockout NOD mice are protected from disease48 while Ins2 knockout mice have accelerated disease.48,49
Insulin autoreactivity is essential for diabetes development in the NOD mouse,50 and approximately 15 CD4 and CD8 T-cell epitopes for insulin have been identified in NOD mice.46 While there are numerous autoantigens targeted by T cells in type 1 diabetes, reactivity to insulin appears to precede reactivity to other major autoantigens such as islet-specific glucose-6-phosphatase catalytic subunit-related protein.50 This suggests that reactivity to insulin is a precipitating event that causes beta cell damage and release of other autoantigen targets. In particular, the peptide Ins B9-23 must be present for diabetes to develop in NOD mice.51 This peptide is recognized by autoreactive CD4 T cells52 and contains Ins B15-23, a target of CD8 T cells in the NOD mouse.53 Ins B9-23 has also been shown to be a target of autoreactive CD4 T cells in type 1 diabetes patients.54 These findings suggest that insulin-targeted antigen therapy has great potential for success in both NOD mice and patients.
Insulin injections,55 oral insulin,56 and aerosolized insulin57 have all been shown to reduce diabetes incidence in NOD mice when treatment is begun at 4–5 weeks of age. Furthermore, administration of insulin-coupled fixed splenocytes can reverse new-onset disease in NOD mice.58 The success of insulin antigen therapy in NOD mice has inspired several clinical trials in humans. The Diabetes Prevention Trial-Type 1 Diabetes (DPT-1) evaluated the ability of parenteral administration of insulin in at-risk subjects to prevent or delay overt diabetes.59 The subjects were relatives of patients with type 1 diabetes and were considered at high risk for diabetes because of the presence of islet cell antibodies and abnormal glucose tolerance test results. Over a median period of 3·7 years, no delay or prevention of diabetes was observed. A separate DPT-1 trial tested oral insulin administration in relatives of patients with type 1 diabetes who had an intermediate risk of developing the disease (positive for islet cell and insulin autoantibodies but normal glucose tolerance).60 Over a median period of 4·3 years, there was no prevention or delay of disease with this treatment. However, hypothesis-generating subgroup analysis revealed that, in subjects with higher (≥ 80nU/ml) insulin autoantibodies, oral insulin therapy did show a beneficial effect, with a disease delay of 4·5 years. More recently, the Type 1 Diabetes Prediction and Prevention (DIPP) study examined the use of insulin administered intranasally to children with predicted risk because of their HLA genotype and autoantibody levels, but no beneficial effect was observed.61
While none of these trials was ultimately successful, they should not be considered failures. The oral insulin trial provided evidence for an effect in one patient subgroup, and a phase III trial to investigate this further is currently in progress (NCT00419562). Also, the trials generated valuable data that could help to evaluate whether adjustments in parameters such as dose, dose frequency, route of administration, and time of intervention could improve the outcome. Indeed, the ongoing pre-Primary Oral/intranasal INsulin Trial (pre-POINT) will study islet autoantibody-negative children having a high genetic risk for type 1 diabetes and will utilize two different routes of insulin administration (oral or intranasal) and multiple different doses.62 The conditions to be used in the subsequent disease prevention POINT study will be those that elicit an immune response having ‘characteristics consistent with protection’ in the pre-POINT study.62
Studies in NOD mice have suggested possible ways for making insulin antigen therapy more successful. Insulin coupled to the B subunit of cholera toxin (CTB) has been shown to prevent islet infiltration in NOD mice when administered orally in small doses.63 CTB aids in passage across the intestinal barrier, thereby reducing the necessary dosage. Our laboratory has shown that targeting peptides derived from islet autoantigens to steady-state dendritic cells (DCs) can induce tolerance of the corresponding autoreactive T cells in NOD mice.64 This is achieved through the use of an antibody to DEC-205, an endocytic receptor expressed on DCs, which is covalently linked to the antigenic peptide. We are currently working to use this method to deliver insulin peptides and protein to steady-state DCs in NOD mice. Transgenic mice expressing human genes such as human insulin and human MHC susceptibility alleles are also being employed to permit the NOD model to more closely mimic the human disease. Finally, administration of insulin in combination with GAD65 has shown promise in protecting NOD mice from diabetes.65 Indeed, the Immune Tolerance Network–Juvenile Diabetes Research Foundation Type 1 Diabetes Combination Therapy Assessment Group recently advocated the exploration of combination therapies, including administration of two different autoantigens.66
Recently identified NOD T-cell antigens
Hsp60, GAD65, and insulin were all identified as T-cell antigens in NOD mice in the early 1990s and were among the first T-cell targets to be identified in this model (Table 1).6,26,67 Thus, they have had the greatest amount of time to be tested in preclinical models and to transit to human trials. In this section, we will highlight several recently identified NOD T-cell antigens whose translational potential has yet to be explored.
Regenerating gene II (Reg II)
Reg II is one of the most recently identified autoantigens in the NOD mouse.68 It is a member of the regenerating gene (Reg) family whose founding member was identified from a cDNA library prepared from rat islets undergoing regeneration in response to a 90% pancreatectomy and treatment with a poly(ADP-ribose) synthetase inhibitor.69 Reg II was explored as a possible autoantigen in NOD mice not only because it was found to be expressed in beta cells,68 but also because other Reg family members were known to be T-cell antigens in NOD mice70 or the target of autoantibodies in patients with type 1 diabetes.71 When NOD mice were immunized with the N-terminal fragment of Reg II in alum, diabetes was accelerated, suggesting the presence of Reg II epitopes capable of stimulating pathogenic beta cell-specific T cells in NOD mice.68 However, the therapeutic potential of the protein is suggested by the finding that administration of full-length Reg II or its C-terminal fragment delayed disease.68
Pancreatic duodenal homeobox 1 (Pdx1)
The transcription factor Pdx1 is a newly identified autoantigen in NOD mice.72 Pdx1 autoantibodies were found in the serum of NOD mice as young as 5 weeks of age, but were not detectable in C57BL/6 or BALB/c mice. The immunodominant B-cell epitope resides in the C-terminal portion of the protein (amino acids 200–283). Splenic T cells from NOD mice proliferated in response to this fragment of the protein, suggesting that a T-cell epitope is also located within this region, but further study is warranted. In this same report, type 1 diabetes patients and at-risk individuals were shown to harbour autoantibodies to Pdx1, and two immunodominant epitopes were suggested (residues 159–200 and 200–283). Whether Pdx1 is targeted by T cells in type 1 diabetes patients remains to be investigated.
Chromogranin A
Chromogranin A is the newest addition to the list of diabetes-related NOD mouse T-cell antigens.73 It was recently identified as the target for the highly diabetogenic NOD-derived CD4 T-cell clone BDC-2·5,74 whose specificity had remained elusive for over 20 years. Chromogranin A is not specific to the beta cell but rather is found in the secretory granules of a variety of endocrine cells and neurons and is processed into multiple peptides by prohormone convertase enzymes.75 The epitope for BDC-2·5 was found to be one of these naturally processed peptides, designated WE14.73 The peptide binds in a non-traditional manner to the class II MHC molecule I-Ag7, leaving empty the N-terminal half of the peptide-binding groove. The authors of this work speculate that WE14 may be post-translationally modified in vivo (as a partially purified beta cell secretory granule protein preparation stimulates BDC-2·5 better than does synthetic WE14) and that a pancreas-specific modification could explain why BDC-2·5 mediates beta cell autoimmunity while apparently sparing other endocrine cells that also express chromogranin A. Multiple laboratories are undoubtedly currently investigating whether chromogranin A is also an autoantigen in type 1 diabetes patients.
The diverse benefits of antigen identification
The NOD mouse can be used not only to identify new antigens targeted in the disease, but also to evaluate alternate methods for therapeutic antigen delivery once important antigens are discovered. From recent discussions of therapeutic strategies being explored in NOD mice,5,76,77 it can be seen that diverse forms of antigen have been considered (Fig. 2). These include proteins,25 peptides or altered peptide ligands,78,79 antigens linked to cholera toxin B,63 cell-bound antigens (e.g. peptide-pulsed immature DCs80 or antigen-coupled fixed splenocytes58), multimerized peptide/MHC complexes (e.g. class II MHC dimers,81 class I MHC-coated nanoparticles,82 or toxin-coupled class I MHC tetramers83), and antigen-linked antibody molecules (e.g. peptide-linked anti-DEC-20564). While here we have discussed NOD T-cell antigens primarily from the point of view of their value in the development of antigen-specific therapeutic strategies, it should be noted that multiple other benefits derive from antigen discovery. Examples are the ability to image the islet infiltrate in vivo,84 monitor beta cell autoimmunity in type 1diabetes patients and islet transplant recipients using T-cell-based assays,85,86 and gain new insights into disease pathogenesis.73
Figure 2.

T-cell antigen delivery methods employed in non-obese diabetic (NOD) mice. In addition to the administration of free proteins or peptides, a variety of other antigen delivery strategies have been utilized in NOD mice. These are illustrated schematically and are not drawn to scale. See text for referenced examples of each delivery strategy.
Conclusions
Of the 18 known diabetes-relevant T-cell antigens in NOD mice, 12 are also targeted by T cells in patients with type 1 diabetes.5 This illustrates the value of the NOD mouse model in the identification of antigens relevant to the human disease. Indeed, as we have discussed here, three NOD T-cell antigens (Hsp60, GAD65 and insulin) continue to be evaluated in a series of ongoing clinical trials. Furthermore, the usefulness of identified antigens for immune monitoring in the context of intervention trials will increase in the future as T-cell assays for autoreactive specificities in patients continue to improve. The recent identification of several novel T-cell antigens in NOD mice indicates that the model continues to represent fertile ground for antigen discovery research.
Acknowledgments
Related research by our laboratory 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. J. B. was supported by the National Institutes of Health Molecular Neuropathology Training Grant NS07098. A.G. was supported by a Howard Hughes Medical Institute Research Training Fellowship.
Disclosures
The authors have no potential conflicts of interest to declare regarding this article.
References
- 1.Anderson MS, Bluestone JA. The NOD mouse: a model of immune dysregulation. Annu Rev Immunol. 2005;23:447–85. doi: 10.1146/annurev.immunol.23.021704.115643. [DOI] [PubMed] [Google Scholar]
- 2.Ikegami H, Makino S, Harada M, Eisenbarth GS, Hattori M. The cataract Shionogi mouse, a sister strain of the non-obese diabetic mouse: similar class II but different class I gene products. Diabetologia. 1988;31:254–8. doi: 10.1007/BF00290594. [DOI] [PubMed] [Google Scholar]
- 3.Makino S, Kunimoto K, Muraoka Y, Mizushima Y, Katagiri K, Tochino Y. Breeding of a non-obese, diabetic strain of mice. Exp Anim. 1980;29:1–13. doi: 10.1538/expanim1978.29.1_1. [DOI] [PubMed] [Google Scholar]
- 4.Driver JP, Serreze DV, Chen YG. Mouse models for the study of autoimmune type 1 diabetes: an NOD to similarities and differences to human disease. Semin Immunopathol. doi: 10.1007/s00281-010-0204-1. (in press) [DOI] [PubMed] [Google Scholar]
- 5.Chaparro RJ, DiLorenzo TP. An update on the use of NOD mice to study autoimmune (type 1) diabetes. Expert Rev Clin Immunol. doi: 10.1586/eci.10.68. (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Elias D, Markovits D, Reshef T, van der Zee R, Cohen IR. Induction and therapy of autoimmune diabetes in the non-obese diabetic (NOD/Lt) mouse by a 65-kDa heat shock protein. Proc Natl Acad Sci USA. 1990;87:1576–80. doi: 10.1073/pnas.87.4.1576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Elias D, Reshef T, Birk OS, van der Zee R, Walker MD, Cohen IR. Vaccination against autoimmune mouse diabetes with a T-cell epitope of the human 65-kDa heat shock protein. Proc Natl Acad Sci USA. 1991;88:3088–91. doi: 10.1073/pnas.88.8.3088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Birk OS, Elias D, Weiss AS, Rosen A, van-der Zee R, Walker MD, Cohen IR. NOD mouse diabetes: the ubiquitous mouse hsp60 is a β-cell target antigen of autoimmune T cells. J Autoimmun. 1996;9:159–66. doi: 10.1006/jaut.1996.0019. [DOI] [PubMed] [Google Scholar]
- 9.Elias D, Cohen IR. Peptide therapy for diabetes in NOD mice. Lancet. 1994;343:704–6. doi: 10.1016/s0140-6736(94)91582-2. [DOI] [PubMed] [Google Scholar]
- 10.Abulafia-Lapid R, Elias D, Raz I, Keren-Zur Y, Atlan H, Cohen IR. T cell proliferative responses of type 1 diabetes patients and healthy individuals to human hsp60 and its peptides. J Autoimmun. 1999;12:121–9. doi: 10.1006/jaut.1998.0262. [DOI] [PubMed] [Google Scholar]
- 11.Raz I, Elias D, Avron A, Tamir M, Metzger M, Cohen IR. β-cell function in new-onset type 1 diabetes and immunomodulation with a heat-shock protein peptide (DiaPep277): a randomised, double-blind, phase II trial. Lancet. 2001;358:1749–53. doi: 10.1016/S0140-6736(01)06801-5. [DOI] [PubMed] [Google Scholar]
- 12.Elias D, Cohen IR. Treatment of autoimmune diabetes and insulitis in NOD mice with heat shock protein 60 peptide p277. Diabetes. 1995;44:1132–8. doi: 10.2337/diab.44.9.1132. [DOI] [PubMed] [Google Scholar]
- 13.Raz I, Avron A, Tamir M, Metzger M, Symer L, Eldor R, Cohen IR, Elias D. Treatment of new-onset type 1 diabetes with peptide DiaPep277 is safe and associated with preserved beta-cell function: extension of a randomized, double-blind, phase II trial. Diabetes Metab Res Rev. 2007;23:292–8. doi: 10.1002/dmrr.712. [DOI] [PubMed] [Google Scholar]
- 14.Huurman VA, Decochez K, Mathieu C, Cohen IR, Roep BO. Therapy with the hsp60 peptide DiaPep277 in C-peptide positive type 1 diabetes patients. Diabetes Metab Res Rev. 2007;23:269–75. doi: 10.1002/dmrr.691. [DOI] [PubMed] [Google Scholar]
- 15.Lazar L, Ofan R, Weintrob N, Avron A, Tamir M, Elias D, Phillip M, Josefsberg Z. Heat-shock protein peptide DiaPep277 treatment in children with newly diagnosed type 1 diabetes: a randomised, double-blind phase II study. Diabetes Metab Res Rev. 2007;23:286–91. doi: 10.1002/dmrr.711. [DOI] [PubMed] [Google Scholar]
- 16.Schloot NC, Meierhoff G, Lengyel C, et al. Effect of heat shock protein peptide DiaPep277 on β-cell function in paediatric and adult patients with recent-onset diabetes mellitus type 1: two prospective, randomized, double-blind phase II trials. Diabetes Metab Res Rev. 2007;23:276–85. doi: 10.1002/dmrr.707. [DOI] [PubMed] [Google Scholar]
- 17.Fischer B, Elias D, Bretzel RG, Linn T. Immunomodulation with heat shock protein DiaPep277 to preserve beta cell function in type 1 diabetes - an update. Expert Opin Biol Ther. 2010;10:265–72. doi: 10.1517/14712590903555176. [DOI] [PubMed] [Google Scholar]
- 18.Reizis B, Eisenstein M, Bockova J, Konen-Waisman S, Mor F, Elias D, Cohen IR. Molecular characterization of the diabetes-associated mouse MHC class II protein, I-Ag7. Int Immunol. 1997;9:43–51. doi: 10.1093/intimm/9.1.43. [DOI] [PubMed] [Google Scholar]
- 19.Suri A, Walters JJ, Gross ML, Unanue ER. Natural peptides selected by diabetogenic DQ8 and murine I-Ag7 molecules show common sequence specificity. J Clin Invest. 2005;115:2268–76. doi: 10.1172/JCI25350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Elias D, Meilin A, Ablamunits V, Birk OS, Carmi P, Konen-Waisman S, Cohen IR. Hsp60 peptide therapy of NOD mouse diabetes induces a Th2 cytokine burst and downregulates autoimmunity to various β-cell antigens. Diabetes. 1997;46:758–64. doi: 10.2337/diab.46.5.758. [DOI] [PubMed] [Google Scholar]
- 21.Nussbaum G, Zanin-Zhorov A, Quintana F, Lider O, Cohen IR. Peptide p277 of HSP60 signals T cells: inhibition of inflammatory chemotaxis. Int Immunol. 2006;18:1413–9. doi: 10.1093/intimm/dxl074. [DOI] [PubMed] [Google Scholar]
- 22.Zanin-Zhorov A, Cahalon L, Tal G, Margalit R, Lider O, Cohen IR. Heat shock protein 60 enhances CD4+ CD25+ regulatory T cell function via innate TLR2 signaling. J Clin Invest. 2006;116:2022–32. doi: 10.1172/JCI28423. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 23.Baekkeskov S, Aanstoot HJ, Christgau S, et al. Identification of the 64K autoantigen in insulin-dependent diabetes as the GABA-synthesizing enzyme glutamic acid decarboxylase. Nature. 1990;347:151–6. doi: 10.1038/347151a0. [DOI] [PubMed] [Google Scholar]
- 24.Braun M, Ramracheya R, Bengtsson M, Clark A, Walker JN, Johnson PR, Rorsman P. Gamma-aminobutyric acid (GABA) is an autocrine excitatory transmitter in human pancreatic β-cells. Diabetes. 2010;59:1694–701. doi: 10.2337/db09-0797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kaufman DL, Clare-Salzler M, Tian J, et al. Spontaneous loss of T-cell tolerance to glutamic acid decarboxylase in murine insulin-dependent diabetes. Nature. 1993;366:69–72. doi: 10.1038/366069a0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Tisch R, Yang XD, Singer SM, Liblau RS, Fugger L, McDevitt HO. Immune response to glutamic acid decarboxylase correlates with insulitis in non-obese diabetic mice. Nature. 1993;366:72–5. doi: 10.1038/366072a0. [DOI] [PubMed] [Google Scholar]
- 27.Zekzer D, Wong FS, Ayalon O, Millet I, Altieri M, Shintani S, Solimena M, Sherwin RS. GAD-reactive CD4+ Th1 cells induce diabetes in NOD/SCID mice. J Clin Invest. 1998;101:68–73. doi: 10.1172/JCI119878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Petersen JS, Karlsen AE, Markholst H, Worsaae A, Dyrberg T, Michelsen B. Neonatal tolerization with glutamic acid decarboxylase but not with bovine serum albumin delays the onset of diabetes in NOD mice. Diabetes. 1994;43:1478–84. doi: 10.2337/diab.43.12.1478. [DOI] [PubMed] [Google Scholar]
- 29.Pleau JM, Fernandez-Saravia F, Esling A, Homo-Delarche F, Dardenne M. Prevention of autoimmune diabetes in nonobese diabetic female mice by treatment with recombinant glutamic acid decarboxylase (GAD 65) Clin Immunol Immunopathol. 1995;76:90–5. doi: 10.1006/clin.1995.1092. [DOI] [PubMed] [Google Scholar]
- 30.Tian J, Atkinson MA, Clare-Salzler M, Herschenfeld A, Forsthuber T, Lehmann PV, Kaufman DL. Nasal administration of glutamate decarboxylase (GAD65) peptides induces Th2 responses and prevents murine insulin-dependent diabetes. J Exp Med. 1996;183:1561–7. doi: 10.1084/jem.183.4.1561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Tian J, Clare-Salzler M, Herschenfeld A, et al. Modulating autoimmune responses to GAD inhibits disease progression and prolongs islet graft survival in diabetes-prone mice. Nat Med. 1996;2:1348–53. doi: 10.1038/nm1296-1348. [DOI] [PubMed] [Google Scholar]
- 32.Tisch R, Liblau RS, Yang XD, Liblau P, McDevitt HO. Induction of GAD65-specific regulatory T-cells inhibits ongoing autoimmune diabetes in nonobese diabetic mice. Diabetes. 1998;47:894–9. doi: 10.2337/diabetes.47.6.894. [DOI] [PubMed] [Google Scholar]
- 33.Tisch R, Wang B, Weaver DJ, Liu B, Bui T, Arthos J, Serreze DV. Antigen-specific mediated suppression of beta cell autoimmunity by plasmid DNA vaccination. J Immunol. 2001;166:2122–32. doi: 10.4049/jimmunol.166.3.2122. [DOI] [PubMed] [Google Scholar]
- 34.Jun HS, Chung YH, Han J, Kim A, Yoo SS, Sherwin RS, Yoon JW. Prevention of autoimmune diabetes by immunogene therapy using recombinant vaccinia virus expressing glutamic acid decarboxylase. Diabetologia. 2002;45:668–76. doi: 10.1007/s00125-002-0806-9. [DOI] [PubMed] [Google Scholar]
- 35.Goudy KS, Wang B, Tisch R. Gene gun-mediated DNA vaccination enhances antigen-specific immunotherapy at a late preclinical stage of type 1 diabetes in nonobese diabetic mice. Clin Immunol. 2008;129:49–57. doi: 10.1016/j.clim.2008.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Maron R, Melican NS, Weiner HL. Regulatory Th2-type T cell lines against insulin and GAD peptides derived from orally- and nasally-treated NOD mice suppress diabetes. J Autoimmun. 1999;12:251–8. doi: 10.1006/jaut.1999.0278. [DOI] [PubMed] [Google Scholar]
- 37.Ma S, Huang Y, Yin Z, Menassa R, Brandle JE, Jevnikar AM. Induction of oral tolerance to prevent diabetes with transgenic plants requires glutamic acid decarboxylase (GAD) and IL-4. Proc Natl Acad Sci USA. 2004;101:5680–5. doi: 10.1073/pnas.0307420101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Tarbell KV, Lee M, Ranheim E, et al. CD4+ T cells from glutamic acid decarboxylase (GAD)65-specific T cell receptor transgenic mice are not diabetogenic and can delay diabetes transfer. J Exp Med. 2002;196:481–92. doi: 10.1084/jem.20011845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Yoon JW, Yoon CS, Lim HW, et al. Control of autoimmune diabetes in NOD mice by GAD expression or suppression in beta cells. Science. 1999;284:1183–7. doi: 10.1126/science.284.5417.1183. [DOI] [PubMed] [Google Scholar]
- 40.Yamamoto T, Yamato E, Tashiro F, et al. Development of autoimmune diabetes in glutamic acid decarboxylase 65 (GAD65) knockout NOD mice. Diabetologia. 2004;47:221–4. doi: 10.1007/s00125-003-1296-0. [DOI] [PubMed] [Google Scholar]
- 41.Jaeckel E, Klein L, Martin-Orozco N, von Boehmer H. Normal incidence of diabetes in NOD mice tolerant to glutamic acid decarboxylase. J Exp Med. 2003;197:1635–44. doi: 10.1084/jem.20030215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Agardh CD, Cilio CM, Lethagen A, et al. Clinical evidence for the safety of GAD65 immunomodulation in adult-onset autoimmune diabetes. J Diabetes Complications. 2005;19:238–46. doi: 10.1016/j.jdiacomp.2004.12.003. [DOI] [PubMed] [Google Scholar]
- 43.Agardh CD, Lynch KF, Palmer M, Link K, Lernmark A. GAD65 vaccination: 5 years of follow-up in a randomised dose-escalating study in adult-onset autoimmune diabetes. Diabetologia. 2009;52:1363–8. doi: 10.1007/s00125-009-1371-2. [DOI] [PubMed] [Google Scholar]
- 44.Ludvigsson J, Faresjo M, Hjorth M, et al. GAD treatment and insulin secretion in recent-onset type 1 diabetes. N Engl J Med. 2008;359:1909–20. doi: 10.1056/NEJMoa0804328. [DOI] [PubMed] [Google Scholar]
- 45.Zhang L, Nakayama M, Eisenbarth GS. Insulin as an autoantigen in NOD/human diabetes. Curr Opin Immunol. 2008;20:111–8. doi: 10.1016/j.coi.2007.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.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]
- 47.Heath VL, Moore NC, Parnell SM, Mason DW. Intrathymic expression of genes involved in organ specific autoimmune disease. J Autoimmun. 1998;11:309–18. doi: 10.1006/jaut.1998.0210. [DOI] [PubMed] [Google Scholar]
- 48.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]
- 49.Thebault-Baumont K, Dubois-Laforgue D, Krief P, et al. Acceleration of type 1 diabetes mellitus in proinsulin 2-deficient NOD mice. J Clin Invest. 2003;111:851–7. doi: 10.1172/JCI16584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.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]
- 51.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]
- 52.Daniel D, Gill RG, Schloot N, Wegmann D. Epitope specificity, cytokine production profile and diabetogenic activity of insulin-specific T cell clones isolated from NOD mice. Eur J Immunol. 1995;25:1056–62. doi: 10.1002/eji.1830250430. [DOI] [PubMed] [Google Scholar]
- 53.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]
- 54.Alleva DG, Crowe PD, Jin L, et al. A disease-associated cellular immune response in type 1 diabetics to an immunodominant epitope of insulin. J Clin Invest. 2001;107:173–80. doi: 10.1172/JCI8525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Atkinson MA, Maclaren NK, Luchetta R. Insulitis and diabetes in NOD mice reduced by prophylactic insulin therapy. Diabetes. 1990;39:933–7. doi: 10.2337/diab.39.8.933. [DOI] [PubMed] [Google Scholar]
- 56.Zhang ZJ, Davidson L, Eisenbarth G, Weiner HL. Suppression of diabetes in nonobese diabetic mice by oral administration of porcine insulin. Proc Natl Acad Sci USA. 1991;88:10252–6. doi: 10.1073/pnas.88.22.10252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Harrison LC, Dempsey-Collier M, Kramer DR, Takahashi K. Aerosol insulin induces regulatory CD8 γδ T cells that prevent murine insulin-dependent diabetes. J Exp Med. 1996;184:2167–74. doi: 10.1084/jem.184.6.2167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.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]
- 59.Diabetes Prevention Trial-Type 1 Diabetes Study Group. Effects of insulin in relatives of patients with type 1 diabetes mellitus. N Engl J Med. 2002;346:1685–91. doi: 10.1056/NEJMoa012350. [DOI] [PubMed] [Google Scholar]
- 60.Diabetes Prevention Trial-Type 1 Diabetes Study Group. Effects of oral insulin in relatives of patients with type 1 diabetes: the Diabetes Prevention Trial–Type 1. Diabetes Care. 2005;28:1068–76. doi: 10.2337/diacare.28.5.1068. [DOI] [PubMed] [Google Scholar]
- 61.Nanto-Salonen K, Kupila A, Simell S, et al. Nasal insulin to prevent type 1 diabetes in children with HLA genotypes and autoantibodies conferring increased risk of disease: a double-blind, randomised controlled trial. Lancet. 2008;372:1746–55. doi: 10.1016/S0140-6736(08)61309-4. [DOI] [PubMed] [Google Scholar]
- 62.Achenbach P, Barker J, Bonifacio E. Modulating the natural history of type 1 diabetes in children at high genetic risk by mucosal insulin immunization. Curr Diab Rep. 2008;8:87–93. doi: 10.1007/s11892-008-0017-y. [DOI] [PubMed] [Google Scholar]
- 63.Bergerot I, Ploix C, Petersen J, et al. A cholera toxoid-insulin conjugate as an oral vaccine against spontaneous autoimmune diabetes. Proc Natl Acad Sci USA. 1997;94:4610–4. doi: 10.1073/pnas.94.9.4610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.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]
- 65.Ramiya VK, Shang XZ, Wasserfall CH, Maclaren NK. Effect of oral and intravenous insulin and glutamic acid decarboxylase in NOD mice. Autoimmunity. 1997;26:139–51. doi: 10.3109/08916939708994736. [DOI] [PubMed] [Google Scholar]
- 66.Matthews JB, Staeva TP, Bernstein PL, Peakman M, von Herrath M. Developing combination immunotherapies for type 1 diabetes: recommendations from the ITN-JDRF Type 1 Diabetes Combination Therapy Assessment Group. Clin Exp Immunol. 2010;160:176–84. doi: 10.1111/j.1365-2249.2010.04153.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Wegmann DR, Norbury-Glaser M, Daniel D. Insulin-specific T cells are a predominant component of islet infiltrates in pre-diabetic NOD mice. Eur J Immunol. 1994;24:1853–7. doi: 10.1002/eji.1830240820. [DOI] [PubMed] [Google Scholar]
- 68.Gurr W, Shaw M, Li Y, Sherwin R. RegII is a β-cell protein and autoantigen in diabetes of NOD mice. Diabetes. 2007;56:34–40. doi: 10.2337/db06-0669. [DOI] [PubMed] [Google Scholar]
- 69.Terazono K, Yamamoto H, Takasawa S, Shiga K, Yonemura Y, Tochino Y, Okamoto H. A novel gene activated in regenerating islets. J Biol Chem. 1988;263:2111–4. [PubMed] [Google Scholar]
- 70.Gurr W, Yavari R, Wen L, Shaw M, Mora C, Christa L, Sherwin RS. A Reg family protein is overexpressed in islets from a patient with new-onset type 1 diabetes and acts as T-cell autoantigen in NOD mice. Diabetes. 2002;51:339–46. doi: 10.2337/diabetes.51.2.339. [DOI] [PubMed] [Google Scholar]
- 71.Shervani NJ, Takasawa S, Uchigata Y, et al. Autoantibodies to REG, a beta-cell regeneration factor, in diabetic patients. Eur J Clin Invest. 2004;34:752–8. doi: 10.1111/j.1365-2362.2004.01419.x. [DOI] [PubMed] [Google Scholar]
- 72.Li SW, Koya V, Li Y, Donelan W, Lin P, Reeves WH, Yang LJ. Pancreatic duodenal homeobox 1 protein is a novel beta-cell-specific autoantigen for type I diabetes. Lab Invest. 2010;90:31–9. doi: 10.1038/labinvest.2009.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Stadinski BD, Delong T, Reisdorph N, et al. Chromogranin A is an autoantigen in type 1 diabetes. Nat Immunol. 2010;11:225–31. doi: 10.1038/ni.1844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Haskins K, Portas M, Bergman B, Lafferty K, Bradley B. Pancreatic islet-specific T-cell clones from nonobese diabetic mice. Proc Natl Acad Sci USA. 1989;86:8000–4. doi: 10.1073/pnas.86.20.8000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Zhao E, Zhang D, Basak A, Trudeau VL. New insights into granin-derived peptides: evolution and endocrine roles. Gen Comp Endocrinol. 2009;164:161–74. doi: 10.1016/j.ygcen.2009.01.011. [DOI] [PubMed] [Google Scholar]
- 76.Mukherjee G, Dilorenzo TP. The immunotherapeutic potential of dendritic cells in type 1 diabetes. Clin Exp Immunol. 2010;161:197–207. doi: 10.1111/j.1365-2249.2010.04157.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Shoda LK, Young DL, Ramanujan S, et al. A comprehensive review of interventions in the NOD mouse and implications for translation. Immunity. 2005;23:115–26. doi: 10.1016/j.immuni.2005.08.002. [DOI] [PubMed] [Google Scholar]
- 78.Alleva DG, Gaur A, Jin L, et al. Immunological characterization and therapeutic activity of an altered-peptide ligand, NBI-6024, based on the immunodominant type 1 diabetes autoantigen insulin B-chain (9-23) peptide. Diabetes. 2002;51:2126–34. doi: 10.2337/diabetes.51.7.2126. [DOI] [PubMed] [Google Scholar]
- 79.Han B, Serra P, Amrani A, Yamanouchi J, Maree AF, Edelstein-Keshet L, Santamaria P. 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]
- 80.Haase C, Yu L, Eisenbarth G, Markholst H. Antigen-dependent immunotherapy of non-obese diabetic mice with immature dendritic cells. Clin Exp Immunol. 2010;160:331–9. doi: 10.1111/j.1365-2249.2010.04104.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Li L, Yi Z, Wang B, Tisch R. Suppression of ongoing T cell-mediated autoimmunity by peptide-MHC class II dimer vaccination. J Immunol. 2009;183:4809–16. doi: 10.4049/jimmunol.0901616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Tsai S, Shameli A, Yamanouchi J, et al. Reversal of autoimmunity by boosting memory-like autoregulatory T cells. Immunity. 2010;32:568–80. doi: 10.1016/j.immuni.2010.03.015. [DOI] [PubMed] [Google Scholar]
- 83.Vincent BG, Young EF, Buntzman AS, Stevens R, Kepler TB, Tisch RM, Frelinger JA, Hess PR. Toxin-coupled MHC class I tetramers can specifically ablate autoreactive CD8+ T cells and delay diabetes in nonobese diabetic mice. J Immunol. 2010;184:4196–204. doi: 10.4049/jimmunol.0903931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Medarova Z, Tsai S, Evgenov N, Santamaria P, Moore A. In vivo imaging of a diabetogenic CD8+ T cell response during type 1 diabetes progression. Magn Reson Med. 2008;59:712–20. doi: 10.1002/mrm.21494. [DOI] [PubMed] [Google Scholar]
- 85.Mallone R, Martinuzzi E, Blancou P, et al. CD8+ T-cell responses identify β-cell autoimmunity in human type 1 diabetes. Diabetes. 2007;56:613–21. doi: 10.2337/db06-1419. [DOI] [PubMed] [Google Scholar]
- 86.Velthuis JH, Unger WW, Abreu JR, et al. Simultaneous detection of circulating autoreactive CD8+ T-cells specific for different islet cell-associated epitopes using combinatorial MHC multimers. Diabetes. 2010;59:1721–30. doi: 10.2337/db09-1486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Elliott JF, Qin HY, Bhatti S, Smith DK, Singh RK, Dillon T, Lauzon J, Singh B. Immunization with the larger isoform of mouse glutamic acid decarboxylase (GAD67) prevents autoimmune diabetes in NOD mice. Diabetes. 1994;43:1494–9. doi: 10.2337/diab.43.12.1494. [DOI] [PubMed] [Google Scholar]
- 88.Karges W, Hammond-McKibben D, Cheung RK, Visconti M, Shibuya N, Kemp D, Dosch HM. Immunological aspects of nutritional diabetes prevention in NOD mice: a pilot study for the cow's milk-based IDDM prevention trial. Diabetes. 1997;46:557–64. doi: 10.2337/diab.46.4.557. [DOI] [PubMed] [Google Scholar]
- 89.Kelemen K, Crawford ML, Gill RG, Hutton JC, Wegmann D. Cellular immune response to phogrin in the NOD mouse: cloned T-cells cause destruction of islet transplants. Diabetes. 1999;48:1529–34. doi: 10.2337/diabetes.48.8.1529. [DOI] [PubMed] [Google Scholar]
- 90.Liddi R, Beales PE, Rosignoli G, Pozzilli P. Incomplete Freund's adjuvant reduces diabetes in the non-obese diabetic mouse. Horm Metab Res. 2000;32:201–6. doi: 10.1055/s-2007-978622. [DOI] [PubMed] [Google Scholar]
- 91.Trembleau S, Penna G, Gregori S, Magistrelli G, Isacchi A, Adorini L. Early Th1 response in unprimed nonobese diabetic mice to the tyrosine phosphatase-like insulinoma-associated protein 2, an autoantigen in type 1 diabetes. J Immunol. 2000;165:6748–55. doi: 10.4049/jimmunol.165.12.6748. [DOI] [PubMed] [Google Scholar]
- 92.Winer S, Tsui H, Lau A, et al. Autoimmune islet destruction in spontaneous type 1 diabetes is not β-cell exclusive. Nat Med. 2003;9:198–205. doi: 10.1038/nm818. [DOI] [PubMed] [Google Scholar]
- 93.Lieberman SM, Evans AM, Han B, et al. Identification of the β cell antigen targeted by a prevalent population of pathogenic CD8+ T cells in autoimmune diabetes. Proc Natl Acad Sci USA. 2003;100:8384–8. doi: 10.1073/pnas.0932778100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Lieberman SM, Takaki T, Han B, Santamaria P, Serreze DV, DiLorenzo TP. Individual nonobese diabetic mice exhibit unique patterns of CD8+ T cell reactivity to three islet antigens, including the newly identified widely expressed dystrophia myotonica kinase. J Immunol. 2004;173:6727–34. doi: 10.4049/jimmunol.173.11.6727. [DOI] [PubMed] [Google Scholar]
