Chapter 3

(4/29/2011)

Animal Models of Type 1 Diabetes:

Genetics and Immunological Function

George S. Eisenbarth

Modified from second web edition Lang J and Bellgrau D

Introduction

The past two decades has seen remarkable advances in understanding the genetics and pathophysiology of spontaneous animal models of immune mediated diabetes (Type 1A) including structural characterization of class II MHC presentation of insulin peptide B:9-231 as well as the chromagranin peptide WE-14 (previously elusive target of the  BDC2.5 T cell receptor2), the creation of new animal models 3 with either genetic manipulation of autoantigens4, 5 or  immunologic function (e.g., toll receptor activation; peptide immunization, mutations of regulatory pathways such as AIRE6 or foxP37), or a combination of genetic and environmental manipulation 8-18. “Humanized” mouse strains are being created and studied and promise additional insights relevant to type 1A diabetes 19-21.  In addition at the Jackson laboratory a repository of type 1 diabetes related mouse models has been created which is an important resource for the long-term preservation of the multiple strains that have been created and a means to facilitate sharing of strains by different research groups. 

 

Spontaneous type 1 diabetes-susceptible models include the non-obese diabetic (NOD) mouse, the BioBreeding Diabetes-Prone (BB-DP) rat, the Komeda Diabetes-Prone (KDP) sub-line of the Long-Evans Tokushima Lean rat Lew.1.WR1 and the Lew.1AR1/Ztm rat. Multiple experimentally-induced models of type 1 diabetes are available including: 1) T cell receptor (TCR) transgenic (Tg) and retrogenic mice with the T cell receptors of naturally occurring diabetogenic clones 2) Neo-antigen (Ag) expression under the control of the rat insulin promoter (RIP) to establish neo-self antigen pancreatic expression that can be the target of autoimmunity, and 3) RIP-driven expression of costimulatory molecules on beta cells.  Mice with knockouts of putative islet autoantigens have allowed direct testing of the pathogenic significance of specific target molecules. Strains of mice with mutations of genes associated with type 1 diabetes in man (FoxP3 and AIRE) are being studied (including an autosomal dominant “human” AIRE mutation6).  Such strains usually do not develop diabetes, but rather have novel autoimmune phenotypes 22-24.

 

A major conclusion from these models is that type 1 diabetes is not the result of a single pathway but can be the result of numerous distinct mechanisms 25. Nonetheless, some generalities do exist including the polygenic nature of the disease, the involvement of T cells in the destruction of pancreatic beta cells, and the incomplete penetrance of the disease implying that environmental or “stochastic” factors influence disease susceptibility. These models provide useful tools for studying the autoimmune process as well as testing potential treatments for the prevention of type 1 diabetes. There has been some pessimism26, appropriately expressed 10, that therapies effective in animal models such as the NOD mouse may not be directly relevant to man. No doubt the usual animal models are inbred (thus not diallelic at any locus) and finely “balanced” in terms of progression to diabetes while raised in pathogen-free environments. Thus, it is likely that many more therapies will be effective in the animal models compared to man (Entelos has developed a computer model of NOD mice and catalogued therapeutics27). Nevertheless, the initial experimental success of anti-CD3 monoclonal antibody therapy in man to delay loss of insulin secretion is a direct result of studies in the NOD mouse 28, and large trials such as the Diabetes Prevention Trial (DPT) have provided preliminary data that oral insulin (also first studied in the NOD mouse) in subjects with elevated insulin autoantibodies may delay disease progression to diabetes29. We suspect that informed optimism relative to the utility of animal models is in order26 , and that the more robust the therapy in the animal model (e.g., ability to abrogate insulitis, ability to prevent diabetes in models engineered to be more highly penetrant [e.g., insulin 2 gene knockout NOD mice 30) , reversal of hyperglycemia, and the closer the human studies mimic the animal studies (e.g., dose and timing), the more relevant the study will be to human type 1 diabetes.

 

The Nonobese Diabetic Mouse

            There are now more than 6,000 articles listed in PubMed on the NOD mouse making any comprehensive review of the model a difficult task with the certainty of failure to cite all important contributions and a certainty that not all the different pathways being pursued will be covered.  Given the ability to electronically access primary articles through the internet with services such as PubMed and Google my purpose in this review on such a large topic is to provide an overall organization, a particular viewpoint (with caveats), and to attempt to highlight many important “relevant” observations.  The general hypothesis my laboratory group is pursuing is that NOD mice 1. have relatively mild defects of immune tolerance (particularly in comparison to foxP3 or AIRE mutant mice) and thus the polygenic nature of the disease and the small influence of most loci; 2. that the disease results because of an immune response that once directed at a specific beta cell peptide is essentially normal, though the targeting leads to disease and there is spreading of autoimmunity, with implication that all mechanisms available to the normal immune system for destruction are operative; 3. that there is a primary autoantigenic epitope presented by the class II I-Ag7 molecule and for the NOD mouse it is the insulin B:9-23 peptide presented in “register 3” of I-Ag7 31 and recognized primarily by a conserved alpha chain (TRAV5D-4) of the T cell receptor32 and 4. The killing of islet beta cells is asynchronous with different islets destroyed over time until enough destruction has occurred for hyperglycemia to develop.  These “biases” are stated, as to some extent they influence “commissions” and “omissions” of this review that many investigators will hopefully correct in the future.

Figure 1. Derivation of the spontaneous animal model of type 1A diabetes, the NOD mouse.

The NOD mouse is the most-studied animal model of type 1A diabetes33-37;26, 27, 38-51. The NOD inbred mouse line was established at the Shionogi Research Laboratories in Japan through sib breeding of a hyperglycemic female mouse from the CTS sub line 52 (Figure 1). The line that actually became the NOD mouse strain was originally a control line for the modestly hyperglycemic line that became the NON strain (Non-obese Non-diabetic). NOD mice show islet infiltration by lymphocytes around 5-7 weeks of age followed by the spontaneous development of overt diabetes in approximately 70% of the females and 40% of the males by 30 weeks of age 53, 54. Similar to humans, NOD mice usually (but not always) express anti-insulin autoantibodies in their serum prior to hyperglycemia 55, 56;57 (Figure 2).  Of note NOR mice which do not usually develop diabetes follow the same age dependent pattern of expression of insulin autoantibodies.  NOR mice are a complex congenic inbred strain with approximately 80% NOD genome and following depletion of CD4+CD25+ T cells NOR splenocytes can mediate diabetes in NOD-scid mice58.

 

Figure 2. Insulin autoantibodies and glucose of individual NOD mice followed from 4 weeks of age.

Although all NOD mice develop insulitis, this is not always followed by diabetes. Diabetes incidence varies among established NOD colonies, although in all cases, unlike in humans, gender influences disease incidence with higher disease frequencies in female mice than males 54. Type 1 diabetes incidence is also affected by environmental conditions with higher disease frequencies in sterile conditions 59 and lower disease penetrance with infectious agents 60, 61. The disease can be prevented in numerous ways (>134) 26 including immunological or genetic manipulations of NOD mice 62.

 

Diabetes susceptibility in the NOD mouse results from the interaction of multiple genes63, with the strongest predisposing effect deriving from genes within the major histocompatibility complex (MHC) 64-66. Multiple studies suggest that the effect of the MHC 64 is due to the combination of the unique sequence of the class II I-Ag7 allele with its beta chain lacking aspartic acid at position 57 and proline at position 56 67 (the I-Aalpha chain sequence of NOD is identical to BALB/c), a lack of expression of I-E 68 due to a common mouse mutation (also present in C57BL/6 mice) and specific class I alleles 69, with additional polymorphisms of other genes within the MHC(e.g. idd1670, 71.  In general it has been assumed that the lack of aspartic acid at position 57 of the I-Ag7 beta chain creating a basic I-Ag7 pocket 9 would favor the binding of amino acids with negatively charged side chains into pocket 9.  The recognition of these peptides by the CD4 T cells then creates class II MHC mediated diabetes susceptibility.  Kappler and coworkers have recently discovered that for the insulin peptide B:9-23, just the opposite occurs.  A given peptide can bind to I-Ag7 in different registers with side chains of different amino acids of the peptide binding in pockets 1, 4, 6 and 9 depending on where the peptide docks along the linear I-Ag7 groove (register).  T cell receptors then recognize the peptide MHC complex in a specific register.   For each specific register that a single peptide binds in, the amino acid chains facing the T cell receptor are different.  To go from one register to another, the peptide must rotate for specific side chains to bind in pockets 1,4,6 and 9.   By covalently coupling the B:9-23 peptide in the groove of I-Ag7 with the arginine (peptide amino acid 22) in pocket 9 (unfavored basic amino acid in a basic pocket) it was found that all of the studied anti-B:9-23 autoreactive T cells reacted only when the peptide was in this low affinity register1.  This has led to the hypothesis that such unusual recognition may allow anti-B:9-23 T cells to avoid thymic deletion in that the concentrations of insulin in the thymus are very low.  T cells should be able to recognize the peptide in a low affinity register in the islets where the concentrations of insulin and presumably the B:9-23 peptide presented by I-Ag7 are huge.  Of note Unanue and coworkers have provided data that the B:9-23 peptide is likely created within islets, potentially within insulin secretory granules with subsequent uptake by antigen presenting cells72.

 

A great deal of effort has been involved in mapping the diabetes-associated alleles in both NOD mice and humans. In both cases, numerous loci have been identified (>50), although to date,  few predisposing genes are identified for the NOD mouse (MHC genes 64, potentially CTLA-4 73, IL263, 74 and beta-2 microglobulin important exceptions 66, 75, 76). Genetic analysis has involved mapping of the mouse genome usually with crossing of loci from nonautoimmune-prone strains onto the NOD background as well as analysis of recombinant and related strains. Loci (identified as Iddn in the mouse and IDDMn in the human) with significant LOD scores for association have been analyzed for candidate genes. Multiple congenic strains containing Idd locus (n) from disease-resistant strains have been generated and analyzed for disease incidence, insulitis, and  immunological phenotypes (e.g., insulin autoantibodies 77) associated with diabetes in the NOD mouse 76, 78, 79. A general finding is that several identified regions contain multiple-disease associated loci which alone rarely confer protection but in multitudes of two or more can nearly completely block disease 76. Positional cloning of numerous Idd loci is underway with potential for success of identifying “causative” polymorphisms likely to be dependent upon the number of genes within a given locus. For example a 1.52Mb region of locus Idd5.2 contains 45 genes, while the locus Idd5.1 (2.1-Mb region) contains four genes, including CTLA-4 63, with evidence for influence of CTLA-4 polymorphisms influencing disease in man and mouse.

 

A list of many of the multiple identified Idd loci, their positions on mouse chromosomes, identified phenotypes, and candidate genes is presented in Table 3.1 and a more comprehensive list is provided by Serreze and coworkers 37. These loci can either confer susceptibility or resistance to disease development and even some alleles of the NOD mouse confer resistance rather than susceptibility. Given the identification of these loci, it is possible to rapidly introduce loci from other strains onto the NOD background to create what has been termed speed congenics. Usually within five generations of backcrossing, essentially all NOD diabetogenic loci can be fixed with the locus of interest introduced 80. Though there are reports of some success, a major disadvantage of the NOD mouse is the lack of embryonic stem cell lines to directly create gene knockouts on the NOD background, a lack that is partially alleviated with the availability of “mixed” NOD strain embryonic cell lines 81.

Table 3.1 Loci of the NOD mouse (idd)

Locus

Chromosome

NOD Allele

Phenotype protective allele

Disease Protection

Genes/ (Candidates)

Reference:

idd1

17

Susceptible

No Diabetes

But some

insulitis

100%

MHC class I and II

I-Ag7

Hattori 1986

Todd 1988

Ikegami 2004

Serreze

idd2

9

Susceptible

 

 

 

Wicker 1995

McAleer 1995

idd3

3

Susceptible

Moderate IAA,

Insulitis

69%

(IL2, IL21,Fgf2,Cetn4)

Podolin 1997

Hill 2000

Lyons 2000

Ikegami 2003

Rabinow 2008

idd4

11

Susceptible

age of onset

 

 

Wicker 1995

Devidi-.. 2007

idd5

1

Susceptible

Low IAA, Insulitis

45%

 

Colucci 1997

Hill 2000

idd5.1

1 (2.1 Mb)

Susceptible

CTLA-4

Ligand independent splice

26%

(CTLA-4, Icos, Als2cr19, and Nrp2 (neuropilin))

Hill 2000

Wicker 2004

idd5.2

1 (1.52 Mb)

Susceptible

 

0%

(Nramp1, 45 gene interval)

Hill 2000

Wicker 2004

idd6

6

SusceptiblexB10

ResistantxNON

 

 

 

Wicker 1995

McAleer 1995

idd7

7

Resistant

Low TCR, CD8

 

 

Gonzlez 1997

McAleer 1995

idd8

14

Resistant

 

 

 

Wicker 1995

idd9

4

Susceptible

High insulitis and IAA

90%

(Vav3, Cd30,Tnfr2,Cd137)

Lyons 2000

idd9.1

4

Susceptible

 

 

(Jak1, Lck)

Lyons 2000

idd9.2

4

Susceptible

 

 

(Tnfr2)

Lyons 2000

Siegmund 2000

idd9.3

4

Susceptible

 

20%

 

Lyons 2000

idd10

3

Susceptible

 

36%

Fcgr1 ruled out, CD101

Podolin 1998

Ikegami 2003

idd11

4

Susceptible

Marginal zone B cells

62%

 

Brodnicki 2000

idd12

14

Susceptible

 

74%

 

Wicker 1995

idd13

2

Susceptible

Decrease insulitis

100%

β-2 microglobulin

Serreze 1998

idd13a

2

Susceptible

 

38%

 

Serreze 1998

idd13b

2

Susceptible

 

25%

 

Serreze 1998

idd14

13

Susceptible

 

 

 

McAleer 1995

idd15

5

Susceptible

 

 

(Xmv65)

McAleer 1995

idd16

17

Susceptible

 

52%

H-2k

Ikegami 1995

idd17

3

Susceptible

 

 

 

Podolin 1997

idd18

3

Susceptible

 

9%

 

Podolin 1997

Combined Congenics

 

 

 

 

 

 

10/18

3

Susceptible

Low IAA,

Insulitis

38%

(Cfsm, Cd53, Kcna3, Rap 1a)

Podolin 1998

Robles 2003

3/10/18

3

Susceptible

Low IAA,

Insulitis

92%

 

Lyons 2000

Robles 2003

3/10

3

Susceptible

 

93%

 

Podolin 1997

3/5.1/5.2

1/3

Susceptible

Low IAA, Insulitis

97%

 

Hill 2000

Robles 2003

3/10/18/9

3/ 4

Susceptible

 

100%

 

Lyons 2000

10/17

3

Susceptible

 

58%

 

Podolin 1997

3/10/17

3

Susceptible

 

98%

 

Podolin 1997

5.1/5.2

1

Susceptible

Moderate IAA, Insulitis

50%

 

Robles 2003

Table 3.1 IAA= Insulin Autoantibodies, bold genes likely candidate.

 

Idd1 has been mapped to the MHC locus on mouse chromosome 17 54, 64. Although the class II allele within the MHC does not completely explain the association of disease71, 82 to this locus (other non-MHC class II genes within this locus contribute to disease susceptibility including class I alleles 83, 84), the MHC class II locus is the most studied genes within this region 76. The I-Ag7 molecule behaves as a recessive allele, normally required in the homozygous state for diabetes development in the NOD model 64. Of note, both human DQ and mouse I-A diabetes-associated sequences (DQB1*0302 and IAg7) carry a non-aspartic acid (e.g. serine at position 57 of  b-chain) instead of the aspartic acid residue conserved on other mouse strains and on other DQB1 alleles conferring low diabetes risk in humans 65, 85. These findings have suggested that this particular residue may influence diabetes susceptibility, but it is likely that other amino acids within DQ or IA as well as other MHC genes may be important both in humans and mice. In fact, population studies 86 and experiments in transgenic mice 68, 87 have failed to show position 57 alone as a susceptibility factor and in addition support a significant role for alleles at the DRB1 locus in humans and for the corresponding IE molecule in the mice.

 

The NOD mouse lacks surface expression of the I-E molecule and its expression as a transgene prevents diabetes 68, 87, 88. These data must be interpreted with caution since control experiments with transgenic expression of the diabetogenic IAg7 molecule also protect NOD mice from diabetes 89. The Idd1 locus may act as a gene complex with at least two susceptibility loci, I-A and I-E 87 as well as class I and class III loci71, 82. It must be noted that, as with any one identified Idd locus, the combination of the I-A and I-E NOD alleles is not sufficient for diabetes development as CTS mice share the NOD class II alleles but are disease-resistant 90, 91. In addition, the requirement for IAg7 homozygosity is not absolute as models of mice heterozygous for IAg7 have been found to be susceptible to type 1 diabetes 92.

 

Recent crystallography studies of the IAg7 molecule showed this MHC protein is structurally stable. In comparison to other class II molecules, this diabetes-associated allele contains an altered peptide binding groove which is reported to allow more promiscuous binding of numerous peptides 93, 94, potentially related to earlier studies of weak peptide binding 95. Furthermore, diabetes-associated DQ alleles in humans were also found to have similarly altered peptide-binding grooves in crystallography studies (31). One possible role of weak presentation of self-peptides by MHC in autoimmunity could be inefficient thymic deletion. Support for this concept comes from limiting dilution studies which showed a high frequency of autoreactive T cells in IAg7 mice 96. In general though it appears that I-Ag7 functions normally in peptide presentation and has high affinity for a subset of peptides.  The hypothesis that there is some overarching abnormality of the class II molecules associated with autoimmunity is difficult to reconcile with the observation that some MHC haplotypes protect from one autoimmune disease, while enhancing another, such as the DRB1*1501, DQB1*0602 haplotype association with human multiple sclerosis and type 1 diabetes 97.

 

Investigations into the role of MHC on thymic tolerance processes provide evidence for both a lack of thymic deletion in IAg7 homozygous mice and positive selection of regulatory T cells by protective MHC alleles. The CD4+ anti-islet 4.1 TCR transgenic is positively selected by IAg7 but negatively selected with coexpression of other MHC class II alleles 98. In addition, Ridgway and coworkers showed that dosing the IAg7 allele correlates with the degree of insulitis and autoreactive repertoire 99. On the other hand, BDC2.5 TCR CD4+ anti-islet transgenic mice show selection of regulatory subsets by protective alleles 100. Thus, the current data suggest altered MHC alleles result in inefficient thymic deletion and altered positive selection in the NOD mouse. However, there is also evidence for a role of the IAg7 molecule in the peripheral activation of autoreactive T cells 101. A mutated form of the IAg7 molecule replacing the His and Ser at positions 56 and 57 with the more common Pro and Asp residues generates a transgenic MHC molecule known as BALBg7PD. This MHC transgenic mouse mediates positive selection of BDC2.5 TCR transgenic cells, however, these mature T cells are unable to mount an anti-islet response with BALBg7PD antigen-presenting cells (APCs) whereas responses with NOD WT APCs are adequate 102. In conclusion, the Idd1 MHC locus may contribute to type 1 diabetes both in altering thymic selection processes and facilitating activation of autoreactive T cells in the periphery. Several groups including our own have provided evidence that insulin may a primary autoantigen for the NOD mouse and in particular insulin peptide B:9-234, 5, 103-105.  If this insulin peptide is indeed essential for the development of diabetes of the NOD mouse, the manner in which the B:9-23 peptide binds to I-Ag7 may be a crucial determinant of disease, with report of two binding registers (register 1 and 2)105.    Though a crystal structure of the complete trimolecular complex of I-A87 – B:9-23 peptide and relevant anti-B:9-23 receptor is needed, John Kappler’s laboratory has provided evidence that the peptide binds to I-A87 in a very low affinity register (register 3) in terms of TCR recognition1.  The B:9-23 peptide appears to primarily be targeted by a germ-line encoded V alpha chain (TRAV5D-4) with marked variation in TCR alpha CDR3 and multiple TCR beta chain sequences 106, 107.

 

The BDC 2.5 target antigen is a natural endocrine cell processed peptide of chromagranin, WE-14. “Remarkably” the exact site of N-terminal cleavage to produce WE-14 is essential for BDC 2.5 T cell receptor recognition and the I-A87 groove is only partially filled by the WE-14 peptide 2.  A recent manuscript suggests there is an additional totally different chromogranin peptide also reactive with the BDC 2.5 T cell receptor but this study did not yet study clonal BDC 2.5 T cells. 108.

 

Idd2 has been assigned to mouse chromosome 9 and linked to the T lymphocyte marker thy1, although a significant association with diabetes has not been found in all studies 109.

 

Idd3 Progress in gene identification and contribution to disease has been made with the Idd3 locus with major candidate genes encoding cytokines IL2 and IL21 with at present no clear distinction between the two candidates 37. NOD mice congenic for the Idd3 region of chromosome 3 from C57BL/6 mice show a low incidence of diabetes (25% compared to 80%) 110. The Idd3 locus contains the IL2 gene which has a unique glycosylation form in the NOD 111, however, the protein appears to have normal function and the glycosylation difference has been genetically ruled out as contributing to diabetes74. Identified  idd3 candidate genes in what is now approximately a 650Kb congenic region include Tenr, IL2, IL21, and Fgf2, and Cetn4,  83 76 and two genes of unknown function63, 112. There is a lower expression of IL2 and a higher expression of IL21 with the risk NOD locus. A combination of idd1 and idd3 introgressed onto the C57BL/6 strain is not sufficient for the induction of diabetes with C57 background genes 113.  Engineered haploinsufficiency of IL2 similar to low  expression of IL2 of the NOD mouse associated with idd3114 locus results in reduced CD4+CD25+ regulatory T cells and enhanced diabetes74.

 

 Idd4 maps to chromosome 11 and may influence the frequency and severity of insulitis and progression to diabetes 54, 59. In particular, Idd4 homozygosity determines the age of onset of diabetes and Idd2, Idd3, and Idd4 together may accelerate progression to overt diabetes 115. A recent report indicates that NOD mice with deleted  lipoxygenase involved in the production of proinflammatory fatty acids increase the development of diabetes116. In addition constituitive phosphorylation of Stat5 of NOD mice is associated with idd4117.

 

The Idd5 locus is located on chromosome 1 and alone confers 50% protection from diabetes and in combination with Idd3 provides nearly complete protection from infiltration of the pancreas, thyroid, and salivary glands 118. Therefore, these genes are likely to influence tolerance processes in the animal 119. The synergistic effect follows a model of additivity rather than multiple epistasis 120. Two loci have been identified within the Idd5 region which results in delayed onset of disease. Idd5.1 overlaps with the IDDM12 locus in humans (candidate gene is CD152 or CTLA-4) 118 and which has been narrowed to 2.1-Mb containing CTLA-4, ICOS, Als2cr19, and Nrp2, with CTLA-4 being the primary candidate 73, 121 but higher levels on T cells of ICOS on NOD T cells 122.  The susceptible idd5.1 allele is associated with low levels of a CTLA-4 splice variant that lacks the ligand binding domain to CD80/86 123. Idd5 F2 mice show a resistance to gamma-induced apoptosis in the NOD and NOR strains while T cells from C57BL/6 and DBA/2 strains show a “high-apoptosis” phenotype 124. The CTLA-4 candidate gene is intriguing because CTLA-4-/- mice are also resistant to apoptosis 125. The NOD idd5 locus mediates a bone marrow cell derived defect in negative selection of t cells126.  Microarray data have implicated CD55 (decay accelerating factor) and acyl coenzyme A dehydrogenase expression127 associated with idd5.

 

The Idd6 locus contains the NK cell cluster; NOD.Idd6 (containing NK1.1) congenic mice have been shown to have reduced disease incidence 128. The importance of NK cell function in reducing disease in NOD mice has been a story of great interest 128-130, although the complete localization to Idd6 is still unknown. F2 intercrosses between NOD and C57BL/6 mice showed an association between the Idd6 locus on chromosome 6 and decreased proliferation of immature thymocytes in NOD mouse 131, although the contribution of this phenotype to disease is still unknown. Additional phenotypes associated with idd6 loci include downregulation of expression of Toll like receptor 1 and decreased expression of the Lmp gene97, 132.

 

A recent study indicates that the IDD7 locus influences thymic deletion of specific CD8 autoreactive T cells such as AI4133 and this relates to low expression of the chain of the T cell receptor of AI4.CD8 T cells 133.  Little information is available on Idd8 except that this locus appears to be protective in the homozygous state 115.

 

The Idd9 locus on chromosome 4 is now known to contain 3 distinct loci. NOD.B10 Idd9.1/9.2/9.3 triple congenic mice are almost completely protected from diabetes yet still show insulitis, suggesting these loci are involved in regulating autoreactive T cells 110. A change in cytokine production from IFNg to IL4 by infiltrating cells in triple congenic mice compared to wild-type NOD supports this interpretation. The presence of insulitis and salivary gland infiltrates in the Idd9 congenic mice suggests that tolerance defects still exist. Genes of the TNFR superfamily with polymorphisms between NOD and B10 mice include candidate genes CD30 and TNFR2 for Idd9.2 134 and CD137 for Idd9.3 110.

 

The idd9.1 locus is associated with greater development of NK T cells which may promote immunoregulation 135.  Idd11 has been localized to overlap with the  Idd9.1  locus.  It is reported that in Idd9 mice autoreactive T cell accumulate in the pancreatic lymph node133 and has been reported to influence marginal zone B cells but not confirmed with congenics 136, 137.

 

Idd10 and Idd18 are closely linked on mouse chromosome 3. NOD.B610/18 congenic mice show reduced incidence of diabetes (50% vs. 80%) and include candidate genes Csfm, CD53, KCNA3, Nras, and Rap1a 138. Recent studies evaluating the IIS mouse139 and with congenic mapping indicates that Idd10 is not Fcgr1 83 and CD101 that differs in sequence from NOD for IIS and B6 is a candidate. Adding another locus from chromosome 3 to create NOD.B6 3/10/18 congenic mice results in almost complete loss of diabetes as well as insulitis 140. The few animals that do develop diabetes have a delayed-onset.

 

Idd13, like idd5, is involved in the regulation of T cell progression from benign to destructive insulitis. It contains at least two identified loci: idd13a and idd13b, which contains the gene b2-microglobulin 79. The studies of Serreze and coworkers have firmly established a polymorphism of b2-microglobulin as influencing development of NOD diabetes, and it is one of the few established “genes” outside of the MHC 75. The NOD allele is a standard a isoform, and the b isoform differs from the a by one amino acid (alanine instead of aspartic acid at amino acid 85). The b allele does not suppress diabetes in the presence of the a allele, but the b allele cannot restore diabetes development by transgenesis in mice lacking beta-2 microglobulin but the a isoform does 75.  NOR derived idd13 locus increases invariant NKT cells141.

 

“Monogenic Autoimmune Mutations”

In 1926, Schmidt described a patient with Addison’s disease and thyroiditis 142, and eventually several clinical syndromes consisting of multiple autoimmune disorders were recognized 13. A subset of these syndromes develops from single gene mutations; many such single gene mutations now have animal models. In particular, the Autoimmune Polyendocrine Syndrome Type 1 (APS-I) results from a mutation of the AIRE gene and approximately 18% of patients with this syndrome eventually develop type 1 diabetes (usually with Addison’s disease, mucocutaneous candidiasis, and hypoparathyroidism) 13. When this mutation(deletion) is bred onto mouse strains, lymphocytic infiltrates occur, but no diabetes 24, 143, 144. Of note, however, the AIRE mutation appears to have a major role in expression of “peripheral antigens” within the thymus 23. Hanahan coined the term “peripheral” antigens 145-147, as molecules expressed at low levels within the thymus (e.g., rat-insulin-promoter-driven expression in his studies) and it is clear that such expression has a major influence on autoimmunity to the respective molecules. There is one Italian family reported with an autosomal dominant form of APS-I148 and Anderson and coworkers have introduced this mutation into NOD mice6.  An autosomal dominant autoimmune syndrome develops that differs from the phenotype of NOD mice with both AIRE genes knocked out.  In particular the autosomal dominant disease does not result in pancreatitits6.  The dominant negative mutation appears to act by recruiting wild type AIRE away from active sites of transcription and does down regulate expression of peripheral antigens within the thymus.  The AIRE knockout on NOD mice did not accelerate the development of diabetes and combined myd88 knockouts (eliminating major pathways of toll like receptor signaling) or raising mice in a germ free environment did not influence disease149.

 

The IPEX (Immune dysregulation, Polyendocrinopathy, Enteropathy, X-linked) syndrome is another particularly informative autoimmune syndrome, with mutations of the  Foxp3 gene and the homologous scurfin gene  in mice. This mutation results in loss of a major subset of regulatory T lymphocytes (CD4+CD25+), and overwhelming autoimmunity in man, such that children often die as neonates, and immune-mediated diabetes can occur in the first days of life 22, 150. In the scurfy mouse, the disease can be cured with partial T cell chimerism, and the same appears to be true of man with normal T lymphocytes able to regulate the abnormal immune system in a dominant fashion 151.  Mice with the foxP3 (scurfy) mutation die from overwhelming autoimmunity, but T cell receptor transgenics combined with this mutation, allow studies of specific autoimmunity, and accelerated development of diabetes152.  Depending upon the specific T cell receptor, the Fox P3 mutations can  accelerate diabetes development even in  rag-/- mice expressing a single anti-insulin B:9-23 T cell receptor (Jasinski et al unpublished).

 

The more common autoimmune disorders, similar to the NOD mouse, are polygenic in origin. Human and rat studies of type 1 diabetes, multiple sclerosis, collagen-induced arthritis and systemic lupus erythematosus (SLE) have been mapped to the same chromosomal sites, suggesting a common genetic basis 78, 153, 154. However, given the number of loci, some chance overlap is to be expected. The  autoimmune polymorphism of the PTPN22 gene (LYP gene of man)  contributes to multiple autoimmune diseases (type 1 diabetes, rheumatoid arthritis, Graves’ disease, lupus erythematosus) 155-157). The NOD mouse itself develops multiple autoimmune manifestations in addition to type 1 diabetes including thyroiditis, lymphocyte infiltration of the salivary and lachrymal glands 66, and can develop polyneuropathy 158 if B7.2 deficient.  NOD mice have been reported to target “Schwann” like cells surrounding islets 159, and are susceptible to both experimental autoimmune encephalitis (EAE - an animal model for multiple sclerosis) 160 and experimental autoimmune prostatitis 161. Therefore certain Idd genes or combinations thereof  alter the immune system such that tolerance processes are defective for multiple tissues. The form of autoimmunity may then depend on other genetic (e.g., MHC and antigens) or environmental factors. One recent study shows that autoimmunity in the NOD switches from pancreas-specific to destruction of the peripheral nervous system by altering  costimulatory molecules present on dendritic cells 158 and NOD.c3c4 mice with multiple B6 and B10 protective alleles from chromosome 3 and 4 bred onto NOD do not develop diabetes, but develop autoimmune biliary disease 162. Of note, breeding only one of the two chromosomal regions onto NOD produces mice with biliary autoantibodies without liver infiltration 162. In a similar manner, NOD congenics can express anti-insulin autoantibodies with rare progression to diabetes 77. Thus disease targeting is influenced both by MHC alleles and by other polymorphisms in an autoimmune “background”.

 

Immunology of the NOD mouse

            Beta cell destruction in the NOD requires CD4 T cells, CD8 T cells 163-165 and B cells for its spontaneous occurrence 166, and maternal transfer of autoantibodies has also been found to have a role 167.

 

Th17 cells do not apprear critical for initiation of insulitis168.  The NOD mouse allows studies of the progression of the disease including the timing of insulitis and the nature of insulitis, i.e., cytokine production during the course of the disease. Prior to developing sufficient beta cell damage to produce hyperglycemia, a period of insulitis is observed followed by eventual development of hyperglycemia. There remains a debate among investigators whether the beta-cell loss is a gradual or acute process, occurring suddenly once regulation of autoreactive T cells wanes, but we suspect the term “benign” insulitis is a misnomer. More than 90% of untreated female NOD mice lose beta cell mass with time, such that by 52 weeks beta cell mass is 1/5th to 1/10th of normal even for the great majority of female mice that have not progressed to diabetes 169 (and Gianani et al ADA abstract 2008). There is considerable endocrine reserve and mice even with 1/5th of normal beta cell mass can remain non-diabetic. As illustrated in Figure 3, even 8-9 week old NOD mice have lost beta cell mass. Of note, the histology of the NOD pancreas suggests asynchronous destruction of islet beta cells. Within the same mouse, one can find normal islets with beta cells, islets with insulitis and beta cells, and islets where all the beta cells (insulin producing cells) have been destroyed, and only non-beta cells (e.g., alpha cells producing glucagon) remain. Additionally during the phase of active destruction there is evidence of beta cell replication 169, 170 that eventually cannot keep pace with beta cell destruction.  Recent three dimension analysis of beta cell mass over time indicates that beta cells on the periphery of the pancreas are destroyed first with increased size of central islets, followed by their destruction 171.

 

 

Figure 3. Quantitation of beta cell mass, comparing NOD-SCID mice that lack T cells, to loss of beta cell mass in NOD with age.

 

There is now clear evidence that mice can regenerate beta cell mass following acute beta cell destruction but it is not clear if humans have such an ability40, 172.  Anecdotal reports of beta cell proliferation173 followed by analysis of multiple individuals174 suggest that replication of human beta cells is infrequent in adults 175.  A subset of patients with type 1 diabetes retain C-peptide secretion long-term and infrequent scattered beta cells are present in most long-term patients 176, but the great majority have very low c-peptide levels176.  The mechanism related to retention of some beta cell mass in man and in the NOD mouse model is currently unknown.  Herold and coworkers analyzing NOD mice prior to the development of diabetes and then during therapy with both anti-CD3 and regulatory T lymphocytes have concluded that inflammation increases beta cell replication, that most of the recovery of beta cells following therapy is a result of regranulation of degranulated beta cells and following therapy beta cell replication is reduced177 with lower percentage of Ki67+ beta cells post recovery(figure below).

 

In NOD mice, expression of insulin autoantibodies (IAA) is usually noted prior to onset of hyperglycemia 178. In addition, the NOR mice, a strain closely related to the NOD with the IAg7 MHC haplotype as well as other NOD congenic strains 30, 77, develop IAA but with limited or no progression to type 1 diabetes 77. The presence of insulin autoantibodies correlates primarily with insulitis and not simply progression to hyperglycemia. Genetic studies using congenic NOD strains show the expression of IAA antibodies is controlled by multiple loci. 77 In addition, the amount of IAA was found to associate with degrees of insulitis more than disease incidence 77.  There is evidence of additional autoantibodies reacting with islets cell antigens of NOD mice, but in workshops it has been difficult to confirm the presence of such autoantibodies with highly specific fluid phase radioassays179, 180.  It is likely that B-lymphocytes (though insufficient by themselves181 can not only contribute to diabetes through the production of autoantibodies (e.g. evidence that “transplacental” passage of autoantibodies of NOD mice important for disease182) but have additional roles, particularly as antigen presenting cells and for the maturation of CD8 T cells167, 183.  Treatment of  “humanized” CD20 NOD mice by an anti-CD20 monoclonal in clinical use prevents the bulk of diabetes184.

 

It is likely that antigen presentation for the activation of pathogenic T lymphocytes occurs in the pancreatic lymph node with evidence provided by the BDC 2.5 transgenic T cells, where labeling of cells with the dye CFSE indicates initial proliferation in such lymph nodes 185, 186, the finding by Fathman and coworkers that pathogenic cells in the pancreatic lymph node are CD4high 187, and removal of pancreatic lymph nodes at 3 weeks but not 10 weeks prevents diabetes 188. The role of various cell populations in the beta cell damage have been addressed using knockout animals, T cell clones, and adoptive transfer models of the NOD mouse and are discussed in the following chapter. To summarize numerous findings, spontaneous disease requires CD4 and CD8 T cells as well as B cells, which are thought to be involved in antigen presentation. Mathis and colleagues have implicated NK specific transcripts and proportion of NK cells in development of destructive islet autoimmunity 189.

 

The confirmation that lymphocytes are required for the beta cell destruction in type 1 diabetes led to numerous studies on the immune system in NOD mice compared to nonautoimmune-prone strains. A deficiency in the ability of NOD APCs to mount equivalent T cell responses has been reported 190, 191. These NOD APCs have been found to have low CD86 expression 192, and recent studies have reported phenotypic and functional defects in bone-marrow-derived dendritic cells (DCs) in response to GM-CSF 193. Wong et al. have found a deficiency in priming of NOD T cell responses to both endogenous and exogenous antigens 194. Similarly, defects in TCR-mediated signaling resulting in inferior NOD T cell responses have been linked to alterations in p21ras signaling 195. Neonatal CD28 costimulation has been recently found to restore this signaling as well as protect NOD mice from diabetes, suggesting a relationship between this T cell hyporesponsiveness and disease onset 196. These abnormalities in T cell function may relate “lymphopenia” of the NOD mouse with the hypothesis that homeostatic expansion may contribute to autoimmunity 197.

 

Bellgrau and coworkers found a similar T cell hyporesponsiveness among numerous strains of autoimmune-prone mice susceptible to EAE, SLE, rheumatoid arthritis, and autoimmune hemolytic anemia 198. The prevention of diabetes resulting from administration of immune adjuvants 199, 200 or DNA vaccinations to self-antigens 201 may be related to reversing this hyporesponsive immunity observed in the NOD. Further work is required to clarify the mechanism of protection and possible role of a hyporeactive immune response in autoimmunity.

 

One possible mechanism by which a hyporeactive T cell response may contribute to autoimmunity is through defective negative selection in the thymus. Evidence for poor central tolerance in the NOD includes direct demonstration of inferior thymic deletion in NOD compared to BALB/c mice upon administration of anti-TCR antibodies, increased frequency of autoreactive T cell responses to pancreatic antigens 96, 202-205, and  demonstration that epitope spreading in NOD follows a hierarchy from highest affinity TCR:self-antigen interactions to lower affinities suggesting the spreading hierarchy is determined by the extent of negative selection 206. The concept that with disease progression lower affinity TCR reactions are favored is challenged by the studies of Santamaria and coworkers who observe affinity maturation of the TCR utilized by cells targeting the molecule IGRP (islet-specific glucose-6-phosphatase catalytic subunit-related protein), which is the native molecule targeted by anti-NRP (NOD Related Peptide) CD8 lymphocytes which form the majority of T cells infiltrating islets 207-209. Other thymic defects in the NOD include abnormal corticomedullary environments 210, a deficiency in the number of TCR+ CD4-, CD8- populations in NOD mice, and low proliferation of immature thymocytes 131. Whether these phenotypes have relevance to autoimmunity remains to be determined, although the deficiency in TCR+ DN thymocytes does not appear to be related as it is seen in a number of congenic Idd mouse strains which are not diabetic 211.

 

Regulatory Cells

Table 3.2: Summary of Regulatory Cells influencing diabetes

Regulatory Cell

Induction

Comment

Reference

CD4+ TGFbeta

Insulin

oral

Oral insulin delay of diabetes in NOD mice. Did not delay BB rat diabetes

Zhang (1991)

CD4+CD62L+

Anti-CD3

Anti-CD3 treatment induction, TGFbeta dependent mechanism

Belghith (2003)

CD4+CD25+

BDC2.5

TCR Clonal and Mimotope driven delay/prevent diabetes

CD28-/- loss regulation, greater diabetes

Tang (2004)

CD4+CD25+

 (CD69+, CD45RBintCD62Lhigh)

Insulin

B:9-23

Specific 100% prevention diabetes transfer into SCID, in vivo and in vitro B:9-23; Secrete TGF-β, TNF-α

Mukkherjee (2003)

NK T cells

GalCer

Experimental activation with GalCer protects

Sharif (2001)

CD8+alpha

γδ T cells

Insulin

Naso-resp

Administration of insulin by naso-respiratory route (“no degradation”) induces IL10 producing

Harrison (2003)

NK cells

CD3-DX5+ cells

 

Complete Freund’s adjuvant protection

Lee (2004)

Bitypic NK/DC Regulatory cells

Anti-CD40 ligand

Prevent diabetes in RIP-LCMV model

Homann (2002)

 

GAD Transgenic

Anti-GAD TCR

Two anti-GAD TCR transgenics prevent NOD diabetes

McDevitt (2004)

 

It is well established that autoreactive T cells can be controlled by regulatory cells (Table 3.2). There has been a recent tremendous expansion in knowledge concerning regulatory T lymphocytes 11, 212, and in particular, their role in the NOD mouse and the ability to generate large numbers of CD4+CD25+ regulatory T cells213, 214 and prevent diabetes 212. Evidence for the role of regulatory T cells in delaying or preventing overt diabetes are numerous including prevention of adoptive transfer of diabetes by mixing splenocytes 215 from pre-diabetic NOD mice with the disease-inducing splenocytes from diabetic donors 216, induction of regulatory cells dependent upon TGFbeta with anti-CD3 therapy in recent onset NOD mice 217, and the large literature concerning mucosal tolerance with bystander suppression, including ability of oral insulin to delay diabetes of NOD mice 218. It is clear that these regulatory T cells are heterogeneous in nature, and distinct populations have been elucidated 47, 213, 219-222.

 

Related to the initial studies of thymectomy-induced autoimmunity 223, 224, the remarkable human autoimmune phenotype of mutations of the Foxp3 gene (IPEX syndrome-see chapter 8) associated with neonatal immune mediated diabetes 150, the ability of Foxp3 expression to generate CD4+CD25+ regulatory T cells 225 and suppress NOD diabetes 226, and the elegant models inducing deficiencies of these regulatory T cells 223, 227, the CD4+CD25+ T cells have become perhaps the best characterized regulatory T cell of the NOD mouse 11, 212, 228, 229. NOD mice genetically deficient for CD28 expression succumb to diabetes with faster kinetics than wild type NOD mice 230. This accelerated disease incidence was found to be associated with a loss of CD4+CD25+ regulatory T cells. Of note, as  published by Bluestone and coworkers, 107 in vitro expanded FoxP3 expressing Tregs (using the BDC2.5 T cell receptor transgenic (recognizes islet membrane antigen) as proof of principal) reversed new-onset diabetes in 60% of NOD mice. BDC2.5 Treg cells have been demonstrated in the pancreatic islets and may act at this site to suppress disease given the ability of ICOS blockade to accelerate development of diabetes 231. Similar regulatory T cells have been induced by immunization with the B:9-23 insulin peptide as well as a B24-C36 proinsulin peptide 232, 233.

A critical role of NK T cells in protection from diabetes has recently been demonstrated in NOD mice. A deficiency in number and function (IL4 secretion) of CD1-restricted NK T cells has been observed for many years 234, 235, however, the contribution of this phenotype to diabetes development was unclear. Several recent studies demonstrate the protection offered by increased numbers or function of CD1-restricted NK T cells 128-130.  NK T cells show a restricted TCR repertoire with a predominance of Va14/Ja281 chains. These cells are specific for lipid antigens presented in the CD1 MHC molecule. Upon activation, which can be achieved with alpha-galactosylceramide (aGalCer), NK T cells produce large amounts of cytokines, most notably IL-4 and IL-10. The Idd6 locus contains the NKR-P1 gene cluster, which includes the NK1.1 gene. The NK1.1 gene is absent in NOD mice, making NK T cells more difficult to analyze. Carnaud and coworkers demonstrated a contribution of the Idd6 locus to disease incidence as NOD.NK1.1 congenic mice have reduced diabetes incidence which correlated with improved NK cell function 128. The contribution of NK T cell defects to diabetes incidence was also demonstrated in NOD CD1 KO mice which showed an exacerbated disease course. In addition, increasing the numbers of NK T cells in the NOD by transgenic expression of the Va14Ja28 receptor resulted in reduced disease 129. The protection appears to be related to increased IL4 production observed in the pancreas as IL12 or anti-IL4 antibodies were shown to abolish this protection 129. It appears that increasing numbers of NK T cells is not necessary for protection as activation of the existing NK T cells with GalCer alone protects diabetes in CD1-sufficient NOD  (50-52) 130 but not in CD1-deficient NOD mice  (52). This CD1-restricted NK T activating reagent also enhances the survival of transplanted islets 130.

 

NOD mice injected with complete Freund’s adjuvant are dramatically protected from the development of diabetes. Lee and coworkers have recently reported that this protection is abrogated when NK asialo-GM1 cells are depleted and restoring cells expressing CD3-DX5+ restored protection 236.

Harrison and coworkers have studied the administration of insulin by various routes as a means of protecting NOD mice. They find that administration of insulin by the naso-respiratory route protects from diabetes with the induction of CD8+ alpha (TCR gamma delta T regulatory cells that can prevent NOD diabetes). These cells cannot be generated in day 3 thymectomized mice, but are restored with administration of the above gamma delta T cells 233.

 

Singh and coworkers have reported that insulin B:9-23 peptide immunization generates CD4+CD5+ regulatory T cells capable of preventing diabetes 232

 

Homann and coworkers have utilized anti-CD40 ligand blockade and the RIP-LCMV diabetes model to generate cells with the unusual set of markers of both NK cells and dendritic cells, with the finding that these cells can block the development of diabetes 237.

 

Antigenic Targets

A fundamental question relative to the immunopathogenesis of type 1A diabetes (immune- mediated diabetes) is whether there are primary islet autoantigens. It is clear that multiple islet molecules 51 are the target of autoimmunity in man and animal models 238, 239.   In particular, there are  T cell clones 240 whose target antigens are currently unknown  and a list of well-characterized  “specific” targets for T cell clones (e.g., insulin, IGRP, proinsulin, chromagranin) and newly described islet targets (e.g. Pdx- 1 Pancreatic duodenal homeobox) 241as well as target molecules such as heat shock proteins, GAD,  dystrophia myotonica kinase (DMK) regenerating gene II 242,where the antigen is either minimally or not specifically expressed in mouse islets (GAD) or neurendocrine cells243 or widely expressed in multiple tissues 244, 245.

 

In terms of islet autoantibodies of NOD mice, two autoantibody workshops suggest that only insulin autoantibodies can be specifically detected using sensitive radioassays 180, 246. Despite the large number of islet autoantigens, there is increasing evidence that insulin, and in particular, a specific epitope of insulin (the Wegmann insulin B chain amino acids 9-23) may be an essential target in the NOD mouse model 30, 232, 247-249. Both cloned CD4 and CD8 T cells reacting with an overlapping insulin B chain peptide can mediate adoptive transfer of type 1 diabetes 194, 238. MacLaren and coworkers reported that subcutaneous administration of insulin and insulin B chain prevents the development of diabetes in NOD mice, and Weiner and coworkers reported that oral insulin decreased the development of diabetes 250-253. Wegmann and coworkers isolated T lymphocytes directly from islets of NOD mice. The T lymphocytes were stimulated with islets as “antigen” and following the development of a series of clones, the majority was found to react with insulin 238, 247. Of the clones reacting with insulin, more than 90% reacted with insulin peptide B:9-23. These clones were notable for utilizing  conserved T cell receptor V alpha [TRAV5D-4 (AV13S3)] J alpha gene and AJ53 T cell receptor segments, with variation in the junctional region and no apparent conservation of the Vb chain 254, 255. Despite utilization of this dominant Va-chain motif, one of the clones studied recognized insulin peptide B:9-16 and another four B:13-23 256. Administration of the B:9-23 peptide either intranasally without adjuvant or with a single injection in incomplete Freund’s adjuvant protects the majority of NOD mice from progression to diabetes 257. Wong and coworkers have isolated a NOD CD8 clone reacting with insulin peptide B:15-23 and reported a very high frequency of B:15-23 tetramer-positive cells within islets of NOD mice, though the exact percentage remains controversial 194, 258. Follow-up studies suggest that though present early in lesions the percentage of CD8 anti-B:15-23 T cell clones is more limited with a larger population of IGRP (islet-specific glucose-6-phosphatase catalytic subunit-related protein)-reactive CD8 T cells 239. Santameria and coworkers have immunized NOD mice with peptides of IGRP on nanoparticles and induced high affinity CD8+ T cells that reverse diabetes 259. 

 

Autoantigen Gene Knockouts and Retrogenics

Baekkeskov has produced a GAD65 knockout and breeding this gene onto NOD mice does not effect diabetes development.260 Similar studies of knocking out potential islet target molecules such as IA-2 and IA-2 beta similarly did not influence progression to diabetes261, 262.  Lack of immune response to GAD65 did not influence progression to diabetes243, 263 but there is interesting evidence that anti-GAD responses can be protective263. 

 

Vignali and coworkers have produced a series of retrogenics (bone marrow transplants of stem cells with T cell receptors) with T cell receptors targeting islet molecules.  Though anti-GAD T Cell Receptors did not induce disease, anti-insulin (BDC12-4.1T cell receptor) induced delayed diabetes264 (Note anti B:9-23 12-4.4 TCR sequence studied in that manuscript was different from our diabetogenic BDC12-4.4 retrogenics.).  Further studies by Vignali of a large series of anti-GAD TCR retrogenics documented induction of encephalitits and high titer GAD autoantibodies but no insulitis or diabetes 265.  Vignali’s anti-GAD T cell receptor retrogenics included 10E1 that target GAD peptide 524-543, the same target as the 5A anti-GAD CD4 T cell line that, on transfer into  SCID mice, caused diabetes 266but not insulitis of diabetes.  Mouse islets contain almost no GAD and insulitis can be induced in anti-GAD TCR retrogenics in mice only if induced to express GAD in beta cells (GAD-transgenic) 260.  This suggests that presence of GAD65 is irrelevant to the spontaneous development

 

Table 3.3  Autoantigen Knockouts

Knockout

Insulin Autoantibodies

Insulitis

Diabetes

Reference

Insulin 1

Unaffected

Modest Decrease

90% Prevention

Moriyama 2003

 

Insulin 2

Increased

Increased

Accelerated

Moriyama 2003

Boitard 2003

GAD65

 

 

No Effect

Kash 1999

Yamamoto 2004

IA-2

 

 

No Effect

Kubosaki 2004

IA-2beta (Phogrin)

 

 

No Effect

Kubosaki 2004

 

of diabetes of NOD mice but does not address a potential role of GAD67 and the ability of GAD peptide immunization to prevent diabetes. Yoon and coworkers produced five anti-sense GAD transgenic lines 267 with the transgene bred onto the NOD background. Follow-up of these lines indicates that of the two lines with any diabetes suppression, one develops diabetes spontaneously and the other after cyclophosphamide induction (oral communication). Further study of these transgenic lines will be of particular interest relative to the mechanism of disease alteration.

 

The lack of effect of the GAD65 knockout is concordant with studies inducing tolerance to GAD and finding no influence on progression to diabetes 243. Nevertheless  anti-GAD T cell receptor transgenes mice inhibit  development of diabetes 263. For the NOD mouse there is a consensus that immune recognition of GAD is more related to protection than beta cell destruction263.  In addition, at least one human DR4-restricted anti-GAD TCR transgenic targeting islets has been produced that causes insulitis, but not diabetes 268.

 

HSP60 and peptide 277 have not been studied with knockout techniques. Cohen and coworkers have studied in detail the ubiquitous HSP60 molecule and peptide p277-responsive T lymphocytes of NOD mice 269-272. The administration of the p277 peptide has been reported to prevent diabetes, but to have no effect in a study by Atkinson and coworkers 273. The evidence that HSP60/p277 is an islet “autoantigen” is relatively weak, with no demonstration in workshops of specific reactivity and with an alternative hypothesis for its effects in NOD mice related to activation of the innate immune system 245.

 

Insulin as the Primary Autoantigen of the NOD Mouse

 

To date, only knockouts of insulin genes have influenced development of NOD diabetes (Table 3.3). Mice have two insulin genes, and since both insulins are present in islets and both insulins are metabolically active, it is possible to knock out either gene and not develop metabolic forms of diabetes. The insulin 2 gene is the proinsulin gene that is expressed within the thymus. The proinsulin 1 gene is a retroposon, with almost no expression within the thymus. Single insulin 2 gene knockouts (produced by J. Jami) were bred onto NOD mice by our group 30 and Boitard and coworkers 104, 274. The insulin 2 gene knockout greatly accelerates the development of diabetes and increases the levels of insulin autoantibodies. The insulin 1 gene knockout prevents approximately 90% of the development of diabetes of female NOD mice but does not alter the expression of insulin autoantibodies and the majority of mice with the insulin 2 knockout have insulitis, consistent with the subset progressing to overt diabetes. Jaeckel and coworkers using a technology to induce tolerance similar to that which failed to alter progression to diabetes with GAD65 transgene 243, have recently reported that with an insulin 2 preproinsulin transgene with invariant chain promoter, progression to diabetes is markedly decreased. They concluded that insulin is a “key” autoantigen of the NOD mouse but not “essential” 248. Of note, they only utilized an insulin 2 transgene. Insulin 2 differs from insulin 1 for two amino acids (as well as multiple additional polymorphisms in the leader and connecting peptide sequence). One of the amino acids that differ between insulin 1 and insulin 2 is position 9 of the B:9-23 peptide (serine for insulin 2, proline for insulin 1), and we have in vivo evidence that the immune response to the two peptides can be dramatically different 275. A double insulin gene knockout NOD mouse (insulin 1 and insulin 2) with a mutant preproinsulin transgene to prevent metabolic diabetes prevented diabetes4. The dramatic effect of insulin gene knockouts is consistent with multiple studies indicating induction of widespread insulin gene expression prevents diabetes, including expression in bone marrow-derived cells 276, 277.

 

Multiple islet molecules are the target of autoantibodies or T cells in man and animal models41, 240, 278, with the islet Zinc transporter (ZnNt8) a recent addition279.

 

Figure 4. Life table analysis of progression to diabetes of NOD mice with insulin 1 or insulin 2 gene knocked out, for male and female mice. Homozygous knockout mice are represented by the triangles. Heterozygous knockout by the squares, and wild type at the insulin gene by circles. Knocking out insulin 2 accelerates diabetes whereas insulin 1 knockout prevents the majority of progression to diabetes.

 

There is not universal agreement if any of the target molecules are essential for immune mediated beta cell destruction, though studies from the laboratories of Kay5, 280, Boitard104, 274, Jaeckel248 our group and others, strongly implicate immune responses to insulin as a central component of type 1 diabetes of the NOD mouse. Our studies both in man and mouse have concentrated on insulin30.  Kay and coworkers utilizing a transgene driving proinsulin expression with an I-E promoter completely prevent diabetes and of interest abrogate development of IGRP CD8 reactive T cells, and even prevent diabetes in a TCR transgenic targeting IGRP5, 280.  Of note a similar I-E promoter transgene but driving IGRP expression has no effect on progression to diabetes280, and thus they concluded that immune responses to proinsulin are “upstream” of IGRP T cell targeting and crucial for diabetes, and lack of immune response to insulin even prevented diabetes in mice with an anti-IGRP T cell receptor transgene5. 

We have analyzed NOD mice lacking both insulin 1 and 2 genes, with multiple transgenic founders with either a native insulin sequence or a sequence with a single amino acid altered (B16:A replacing B16:Y).  These mice are protected from diabetes and development of insulin autoantibodies and insulitis are markedly decreased4.  Restoring the native B:9-23 sequence with an islet transplant(but not bone marrow transplant) or peptide immunization, or a native proinsulin transgene, restores anti-insulin autoimmunity and generates CD4 T cells able to cause diabetes103. 

 

BioBreeding Diabetes-Prone (BB-DP) Rat

            The first inbred animal model for spontaneous, type 1 diabetes evolved from the discovery of diabetic animals in an outbred colony of Wistar rats maintained at the Bio Breeding Research Laboratories in Ottawa, Canada. Selective breeding led to the first paper on the Bio Breeding (BB) rat in 1975 wherein the clinical description of insulin-dependent diabetes was documented 281. Prevention of diabetes with antiserum to rat lymphocytes suggested an autoimmune pathogenesis for disease 282. Further support for this hypothesis was provided by studies describing the linkage of the disease with the MHC 283. The disease is now known to have a polygenic basis for susceptibility 284, and abnormalities in T cell function have been described 285-288. In comparison to the NOD animal model for type 1 diabetes, the BB model has certain advantages including higher and more uniform disease penetrance and no gender bias as diabetes is observed in >90% of both male and female BB-DP rats when maintained in pathogen-free conditions 289, 290. The time between insulitis and overt diabetes in the BB-DP rat is much shorter than in the NOD mouse, as diabetes develops between 2-4 weeks after insulitis in the rat model. However, the BB-DP model requires a unique lymphopenia (lyp) locus for spontaneous disease which is not present in the NOD mouse or the vast majority of humans with type 1 diabetes 285. This lyp locus is required in the homozygous state for disease and results in a profound loss of peripheral T cells including a >85% reduction in CD8+ T cells and a 50-80% loss of CD4+ T cells. In this section, we will focus on the genetics and immune status of the BB rat, with a special emphasis on the relationship between T cell lymphopenia, T cell function, and disease susceptibility.

 

Derivation of BB Rats

            All BB rat colonies worldwide were derived from a progenitor stock in Ottawa. As there was considerable genetic heterogeneity in the original Ottawa stock, inbreeding at other locations has predictably led to genetically distinct but related, inbred BB rat lines. In a study of 11 different BB rat colonies describing over 20 different BB sub-lines, inbred lines could be classified as belonging to one of four groups distinguished by eight protein markers from at least three linkage groups 291. Selected for sensitivity or resistance to spontaneous diabetes related to presence (BB-DP) of mutation causing severe lymphopenia, BB rats are subdivided into diabetes-prone (BB-DP) or diabetes-resistant (BB-DR) 292. As described later, the BB-DR sub-line  develop diabetes following experimental manipulations that do not induce diabetes in most other rat strains293.

 

Genetics of BB Rats

            It has long been known that diabetes of the BB rat depends upon polymorphisms of the RT1U major histocompatibility complex as well as a “peculiar” severe T cell lymphopenia inherited in an autosomal recessive manner (lyp [lymphopenia] gene in BB rats that is unrelated to the Lymphoid Tyrosine Phosphatase gene (LYP PTPN22) associated with human type 1 diabetes 155) 294, 295. Lymphopenia results from mutation of one of the Immune Associated Nucleotide Related genes (IAN4).  The frameshift mutation of IAN4 is associated with increased apoptosis, mitochondrial dysfunction296, and potentially heightened T cell receptor signaling297.  In the mouse IAN family genes are predominantly expressed in lymphocytes and expression is upregulated with thymocyte differentiation298.

Figure 5. The lymphopenia gene (IDDM1 of rat) of BB-DP rats (Ian gene).

Essentially all rat models of type 1 diabetes have the permissive RT1u MHC haplotype 299(One exception reported in 1990 induced diabetes with thymectomy and sublethal radiation of PVG/c stain rats 300). Linkage of diabetes to the MHC for BB rats was first defined by Colle and coworkers in an analysis of diabetes incidence among F2 animals from a cross of BB-DP with Lewis strain rats 283. Lewis rats are not prone to diabetes and are incompatible with the BB-DP at the MHC. All diabetic F2 animals were shown to be homozygous for the BB-DP MHC which is the susceptible RT1u haplotype.

Multiple BB rat disease-associated loci (iddmn) have been reported 301-303. The lyp locus was mapped to a syntenic region of rat chromosome 4, is designated iddm1, is responsible for the severe T cell loss when present in the homozygous state 285,  (identified as a frameshift mutation of the IAN4 gene 304, 305).

 

Although the contributions of the iddm1 (lyp) and idddm2 (MHC) loci to disease are quantitatively large, they are not, in and of themselves, sufficient for disease. Fixing the RT1u and lyp alleles when breeding the BB-DP rat to diabetes-resistant ACI 294 and PVG 306 strains did not result in diabetes. Therefore, other loci are clearly involved in rendering the animal susceptible to autoimmunity. Reports link iddm4 (now termed iddm14307) with diabetes susceptibility in crosses between disease-resistant Wistar-Furth (WF) and BB-DR animals which do not normally develop spontaneous disease 301. The BB-DR animals are genetically similar to the BB-DP rat yet are resistant to spontaneous type 1 diabetes due to the lack of the iddm1 locus(lymphopenia gene). Thus the BB-DR animals have normal peripheral T cell numbers. However, autoimmune-susceptibility genes are clearly present in the BB-DR as type 1 diabetes can be induced in BB-DR animals with immune manipulations if administered in a short time window of 25-35 days of age. These manipulations include viral infections 308, poly IC injections 309 and depletion of RT6+ T cells along with an environmental trigger 310 and thymectomy311.  One of the most fascinating BB-DR related models is the induction of diabetes with the Kilham rat virus293, 308, 312-314.  The ability of this virus to induce diabetes in BB-DR rats was discovered following the spontaneous infection of BB-DR colony with the virus.  The virus may act without infection of islet beta cells, with evidence that induction of disease follows TLR9 mediated induction of a series of cytokines and in particular IL12p40.  Chloroquine therapy decreases the development of diabetes293. Genetic loci influencing induced diabetes of the BB-DR and related strains have been defined303, 315 with TCR beta(V beta 13) as one candidate gene for IDDM4307, 316. Backcrosses of (WFxDR-BB)F1 to WF rats resulted in approximately one-half of the pups susceptible to type 1 diabetes induction. Furthermore, this susceptibility mapped strongly to the iddm4 locus on chromosome 4 301. This 2.8-cM region on rat Chromosome (Chr) 4 locus contains several major autoimmunity loci including aia2, aia3, and cia3, and it has been assigned to a 2.8cM region proximal to Lyp/Ian4l1 317. These data, as well as studies in the BB/OK strain 302, 318, crosses between BB-DP and non-autoimmune-prone rats 319, and RT1u congenic strains 299 support the contribution of non-MHC loci in general susceptibility to autoimmunity. Two other loci found to be associated with type 1 diabetes in the BB-DP rat, include the diabetes-susceptible iddm3 and diabetes-resistant iddm5 loci 302, 318. 

 

IDDM14 is localized to approximately 2 million bases on chromosome 4 containing the T cell Receptor beta V-gene locus.  This diabtogenic locus is critical for development of diabetes not only for the BBDP and BBDR strains but also for the LEW.1R1 strain where similar to BB-DR rats diabetes is not spontaneous but can induced with agents such as poly-IC.  Resistant strains at IDDM14 are WF, BN, and F344.  Two additional strains that develop autoimmune diabetes (PVG.R8 and KDP) have not formally been evaluated for linkage to IDDM14, but share a region defined by single nucleotide polymorphisms with the susceptible strains, including mutations in several Tcrb-V genes, (e.g. Tcrb-V13)320.  This leads to the hypothesis, not yet proven, that development of diabetes in these strains may be critically dependent upon  a specific T cell receptor Vb sequence.  This would be analogous to the hypothesis that in NOD mice the Valpha sequence TRAV5-D4 associated with T cell receptors targeting insulin peptide B:9-23 may be critical for insulin autoimmunity and diabetes (though in this case the locus is not polymorphic between strains)107.

 

Immune Dysfunction in the BB-DP Rat and Its Relationship to Lymphopenia

Lymphopenia was first defined in 1981 by Jackson and colleagues 321. An early important characteristic distinguishing lymphopenic from nonlymphopenic animals was the absence of the RT6+ subset of peripheral T cells 322, 323. The under-representation of the CD45R+ T cell subset isoform was also documented 324. The absence of RT6+CD45R+ T cells correlated well with the overall reduction in the numbers of peripheral T cells found in the BB-DP. Since thymocytes are both RT6- 322 and 98% CD45R- 325, a plausible role of the lymphopenia gene is to retard T cell maturation at a stage prior to the expression of these peripheral T cell antigens. The RT6+ population is known to contain regulatory function since 1) depletion of the RT6+ population along with poly I:C injection induces type 1 diabetes in BB-DR rats 326, 2) diabetes can be induced by adoptive transfer of BB-DR lymphocytes into athymic nude WAG rats only if the BB-DR lymphocytes are pretreated with anti-RT6 antibodies while cotransfer of RT6+ T cells prevents diabetes 327 and 3) injection of RT6+ cells from BB-DR rats into BB-DP lymphopenic rats prevents their spontaneous development of type 1 diabetes 328. Both CD4+CD25+ and CD4 T cells that express neither CD25 nor Foxp3 have regulatory function329, 330.

 

Studies of the immune function of lyp T cells from BB-DP rats report contradictory results of poor in vitro responses and hyper-activated phenotypes. Early studies report weak proliferative 331 and cytotoxic responses 287 to alloantigen as well as altered CD4-mediated signaling 332. Given the reduced numbers of T cells in the lyp animals, especially in the CD8 compartment, the differences in responses in these bulk read-out in vitro assays may be due to differences in functional T cells present. It is now known that many of the peripheral T cells in the BB-DP are undergoing apoptosis and therefore may not be functional, although present, in these assays. More recent studies suggest that lyp T cells show increased signs of activation ex vivo, including increased expression of CD25 333 and OX40 activation markers 334, increased INFg production 335, and increased mRNA of T cell signaling adapter protein vav 336. The lyp cells also show increased signs of spontaneous proliferation ex vivo with 1) 2X number of cycling cells as determined by increased DNA content 337, 2) a 90% reduction in levels of cell-cycle inhibitor p27kip, increased levels of PCNA and phosphorylated Rb 338, and 3) incorporation of BrdU into >90% of lyp T cells compared to ~30% of normal T cells in a 13-day period 339. Collectively these data suggest that lyp T cells go through DNA synthesis prior to their imminent demise although no studies have investigated progression through mitosis or cell division. The resultant loss of T cells, as opposed to an accumulation, suggests this proliferative state is non-productive.

 

In addition to T cell abnormalities associated with the lyp gene, studies of non-T cell subsets of the BB-DP immune system suggest thymic and APC defects similar to the NOD mouse model. A lower number of splenic DCs with decreased expression of MHC class II and costimulatory ligand CD80 340 as well as  decreased clustering which improves stimulating capacity of DCs is reported 341. In addition, defects in the NK cell population 342 as well as thymic B cells 343 have been observed. The data support an overwhelming defect in peripheral T cell regulation as a mechanism for disease and reports of enhanced proliferative responses to superantigens following in vivo administration 288 support the conclusion of defects in peripheral tolerance in lyp rats. Few studies on thymic tolerance have been reported in the BB-DP model although alterations in thymic medullary 285 and cortical 344 architecture have been associated with thymic tolerance defects were noted.

 

A remarkable phenotype afforded by the lyp mutation is the severely shortened lifespan of the lyp T cells. Although thymic development appears normal in lyp animals with an almost normal distribution of double-positive and CD4+ and CD8+ single-positive subsets which vary depending on background strain 306, peripheral T cell numbers are drastically reduced. The CD8+ T cell subset is nearly absent in the spleen and lymph nodes of lyp animals with a few cells expressing lower levels of CD8 345, suggesting a faster death following thymic selection than CD4+ T cells 346, 347. Tracking the export of T cells from the thymus with fluorescent-labeling, Zadeh and coworkers measured the lifespan of CD4+ lyp T cells to be less than one week 337. In addition, only few labeled thymocytes expressed RT6, which is normally upregulated 1-2 weeks following thymic export. The majority of the labeled thymocytes expressed the Thy1 Antigen which is characteristic of recent thymic emigrants (RTEs) in the rat 337. The thymus of the lyp rat also showed reduced thymic export of RTEs, data which is consistent with increased death of thymocytes in adult thymic organ cultures (ATOCs) from lyp rats 348. Further data supporting an overwhelmingly shortened lifespan of lyp T cells is the rapid loss of T cells from lymph nodes (LNs) and spleen following thymectomy in lyp rats compared to a fairly stable T cell pool in thymectomized normal rats 349, 350.

 

The efficient clearance of apoptotic cells by phagocytosis has made detection of apoptosis difficult in vivo. Death by apoptosis in lyp T cells is rapid in vitro 306, 339 with a majority (~80%) of the cells dying in overnight cultures compared to negligible death (~20%) observed in cultured T cells from nonlymphopenic rats. Advances in detection of apoptotic cells allowed identification of these dying cells in vivo through TUNEL and FITC-Annexin V staining assays 351. Further characterization identified the clearance of the apoptotic T cells in lyp rats in the liver 342. In mice, dying apoptotic CD8+ T cells are cleared in the liver whereas CD4+ T cells remain in the LN even when apoptotic 352. The dying cells were difficult to define as T cells due to their detectable but low expression of TCR, CD8, CD4, B220 and HSA proteins on their surface 342.

 

The vast reduction in RT6+ cells along with a preponderance of Thy1+ cells suggests lyp impacts T cell development at the RTE (Recent Thymic Emigrant) stage. The effects of the lyp mutation appear to be cell autonomous and restricted to bone marrow cells as demonstrated through lyp à normal bone marrow chimeras 353. In addition, transplants of normal thymus into lyp recipients do not reverse the lymphopenia, pointing to the developing T cells themselves harboring the genetic disturbance 354. It is noteworthy that the T cell loss, activation, and death phenotype observed in the BB-DP rat is also observed in lyp congenic rats from diabetes-resistant strains (Fisher-lyp and PVG-lyp) which show T cell lymphopenia but not diabetes 306, 338.

 

            The reduced number and early death of the T cells and their necessary role in type 1 diabetes induction present a paradox in the BB-DP model. Although a lack of regulation in the BB-DP rat is well-documented, less is known about the properties of the effector T cells responsible for the beta cell destruction. There is disagreement whether the short-lived RTEs are responsible for disease, as suggested by disease prevention with adult thymectomy 350. However, the thymectomy is required during a distinct time period for protection – when performed at 8 weeks of age animals still develop diabetes even though the vast majority of RTEs) still undergo rapid death. Combined with newer studies showing antigen activation can rescue lyp T cells from death both in vivo 339 and in vitro 306, another model suggests that higher affinity autoreactive T cells are activated by autoantigen, survive, and serve the effector functions necessary for immune clearance. Isolation and characterization of the effector T cells responsible for beta cell destruction are still necessary to differentiate among these models.

 

Disease Prevention in the BB-DP Rat

As in the NOD mouse model, numerous treatments have been shown to prevent diabetes in the BB-DP rat model. Early immunosuppression to prevent effector T cell function, including T cell depletion with antibodies 355-357, thymectomy 349, 350, 358 or FK506 359, 360 prevents diabetes in the BB-DP rat. Similar to the effects in the NOD model, complete Freund’s adjuvant 361 and viral infections 362, 363 prevent disease in the BB-DP model as well. Reversing the defect in the regulatory population also prevents diabetes. This reversal has been shown to be successful in disease prevention with the addition of RT6+ cells 364-367 or with human TNFalpha, a cytokine which influences regulatory properties of T cells 368 . Although alterations in peripheral tolerance are well-documented in the BB-DP, little is known about alterations in thymic tolerance in this model. However a report that in thymic organ culture of BB-DP thymocytes, coculture with islets, but not thyrocytes, prevents diabetes and reduces insulitis upon adoptive transfer 369. These data suggest diabetes can be reversed with increased thymic tolerance. The non-MHC and non-lyp loci may indeed influence the repertoire of autoreactive T cells and distinguish lyp animals susceptible or resistant to autoimmunity.

BB rats raised in a germ-free environment develop diabetes.  Nevertheless recent studies indicate that treatment of diabetes-prone BB rats with antibiotics which change their GI flora (with correlation with diabetes outcome), especially when combined with a casein free diet are protected from the development of diabetes370.

 

Long Evans Tokushima Lean (LETL) Rat Model (Komeda Diabetes-prone)

 

An additional rat model of spontaneous development of type 1 diabetes was described in 1991 371. Inbreeding of a rat showing signs of diabetes in 1983 resulted in the LETL strain maintained at the Tokushima Research Institute. These rats have the advantage of spontaneous disease incidence without a gender bias or a requirement for lymphopenia. Like the NOD mouse, infiltration of the pancreas, salivary, and lachrymal glands is observed 371. The disease shows a polygenic mode of inheritance372 including a large contribution of the MHC RT-1u haplotype shared with the BB-DP rat 371. Selection of the Komeda Diabetes-Prone rat substrain improved disease penetrance, with 100% insulitis and 70% diabetes by 120 days of age 373. A genome-wide scan of this strain identified a novel non-MHC associated disease locus on rat chromosome 11 which was shown to be essential for development of insulitis 373.  Mutated Cblb alleles when introduced on a non-KDP RT1(u) rat resulted in a low incidence of diabetes and thyroidits, suggesting additional modifier genes influence penetrance374.  An additional Komeda Non-Diabetic (KND) substrain was established which, although genetically very similar to the KDP strain, does not develop type 1 diabetes 375. This strain serves as an important control in both genetic and immunological studies of the KDP rat. This rat model thus most closely resembles human type 1 diabetes in its spontaneous onset, lack of gender bias, lack of lymphopenia, and association with MHC class II. The non-MHC mutation segregating as a recessive is a nonsense mutation of the Cblb gene (Casitas B-Lineage Lymphoma b gene) which is important for T cell regulation 376. Cbl-b is a ubiquitin ligase important for CD28 co-stimulation during T cell activation. Analysis by two groups for polymorphisms of the human Cbl-b gene failed to find an association with human type 1 diabetes 377, 378.  It does not appear that Cbl-b locus contributes to development of type 1 diabetes of man though there is one report of an interaction with CTLA-4377-379.

Figure 6. The Cblb mutation of the Komeda diabetic rat.

 

LEW.1AR1/Ztm- iddm rat

In 2001 Lenzen and coworkers described a new rat model of insulin deficient diabetes in which approximately 20% of the animals (no sex bias) developed diabetes characterized by lymphocytic infiltration of the pancreas from rats that were not lymphopenic 380, 381. Similar to other rat strains with immune-mediated diabetes, these animals had RT1 haplotype with u alleles (RT1.Aa B/Du Cu 380 and is a recombinant RT1 haplotype (RT1r2). In addition to islets, larger pancreatic ducts show mononuclear infiltrates. The islet infiltrates are predominantly CD8 T cells (>50%) with a relatively small percentage of CD4 cells (4%), and many ED1-positive macrophages 381.   Transfer of CD8+ T cells from prediabetic mice (but not CD4+ cells) protects 382.   At early stages of infiltration macrophages predominate383.  Of note non-diabetic rats lacked islet infiltration, with none of the non-diabetic animals having infiltrates! Other organs, including endocrine organs did not have infiltrates. In summary, the LEW1.AR1/Ztm-iddm rat is a novel model of islet destruction in the presence of islet infiltrates that differs from other models in that age-matched animals lacking diabetes did not have infiltrates. This suggests an acute disease process and one with dramatic heterogeneity between rats.

 

TCR Tg (transgenic) and retrogenic Mouse Models of Type 1 Diabetes

Early studies breeding non-pancreatic specific TCR transgenics, either class-II restricted (D011.10 anti-OVA) or class I-restricted (2C anti-Ld) onto the NOD background resulted in no change in disease incidence 384. Although the transgenes showed good allelic exclusion at the beta locus, the alpha locus was leakier allowing endogenous receptor expression. This finding supported selection for pancreatic-specific T cells in mice skewed T cell repertoires with innocuous receptors.

 

A number of T cell clones have been isolated from the spleens of diabetic NOD mice (BDC2.5, BDC6.9) 385, and the pancreata of pre-diabetic 238, 386-388 NOD or RIP-B7.1 NOD transgenic 389 mice and islet-transplanted diabetic 390 NOD mice, and from islets of NOD mice (BDC12-4.1) 238. Both CD4 and CD8 clones have been generated 391 which have varying abilities to induce or suppress insulitis and diabetes 392. The TCR (T Cell Receptor) of many of these clones have been utilized to produce TCR transgenic (Tg) mice on various backgrounds264. A summary of TCR transgenic diabetic mice is listed in Table 3.4.  A major advance has been the development of the technology to produce retrogenics mice107, 264, 393-397 that has rapidly been applied to diabetogenic T cell receptors.

 

Table 3.4 T Cell Receptor transgenic NOD Mice

T Cell Receptor transgenic

Vbeta

Valpha

CD4/CD8

Antigen

Disease Phenotype

D011.10

8.2

Endogenous

CD4

OVA/IAd

No change in diabetes frequency

2C

8.2

3.1

CD8

Ld

No change in diabetes frequency

BDC2.5

4

 

CD4

Chromagranin

Rapid, uniform insulitis at 3 weeks of age; reduced diabetes incidence; higher incidence of diabetes with CD1d deficiency (?NK T cell protection) and in BDC2.5 C57Bl.IAg7 congenic mice which maps to Idd7 resistance in NOD mice; accelerated diabetes on NOD SCID background

BDC6.9

 

 

CD4

Unknown AutoAg on chr 6 in NOD and SWR pancreas

Accelerated diabetes on NOD background

positively selected on I-Ag7

BDC12-4.1

2.1

13.3 (TRAV 5D-4)

CD4

Insulin B:13-23

Positively selected on I-Ag7 but animals lymphopenic;

Diabetes observed on Rag-/- background; Insulitis on

Mixed strain Rag+ background.

4.1-NOD

 

 

CD4

Unknown AutoAg in pancreas

Positively selected with IAg7; negatively selected with

other class II molecules; Kill via Fas. Increased diabetes incidence; same frequency Rag-/- background  - CD8 independent.

8.3-TCRb

8.1

Endogenous

CD8

IGRP-beta cell specific

Accelerated diabetes with reduced period of

benign insulitis; NRP-V7 peptide tetramer for

diabetes prediction in NOD.

8.3-NOD

8.1

Va17/Ja42

CD8

IGRP

Accelerated diabetes with reduced period of

benign insulitis; Kill by Fas exclusively. On rag-/- background get decreased disease incidence - requires CD4 help.

9.33

6

19

CD8

Unknown AutoAg in pancreas

Accelerated diabetes and increased incidence;

requires CD4 help. No diabetes on scid background –

not due to lack of CD4 but rather lack of escape from

negative selection with only one TCR

NOD.AI4

2

8

CD8

?DMK

DMK widely distributed molecule, mimotope evidence re: target; Needs both Kd and Db for killing; accelerated diabetes in absence of CD4 help or B7.1 expression.

GAD206;GAD524

“Retrogenic”

 

 

CD4

GAD524-543 Subset

Vignali GAD reactive TCR “retrogenic” NOD mice with retroviral stem cell gene transfer. No diabetes TCR’s induced encephalitis.

 

 

 

 

 

 

 

The BDC2.5 TCR transgenic mouse was generated from the BDC2.5 CD4 T cell clone (Ag) 398 which Haskins and coworkers have identified targets the WE-14 peptide of chromogranin clones BDC 10.1 and BDC 5.10.3 also targets chromagranin 2. The BDC2.5 TCR transgenic mouse develops a uniform insulitis at 3 weeks of age followed by a low incidence of overt diabetes. Surprisingly, BDC2.5 transgenic mice on the C57BL/6.IAg7 background showed a higher incidence of diabetes than on the NOD background. The difference in disease incidence was mapped to the Idd7 disease-resistance interval in the NOD 66. The BDC2.5 TCR transgene bred onto the NOD.scid background resulted in a much accelerated onset of type 1 diabetes, suggesting regulatory cells prevent diabetes induction in the normal NOD setting. Of note, diabetes is restored in the absence of NKT cells with deletion of the CD1 locus 399.   BDC2.5 T cells have been utilized to create potent regulatory T cells that are able to suppress the spontaneous disease of the NOD mouse213, 400. 

 

Another CD4+ clone, BDC6.9, isolated in Kathryn Haskin’s laboratory has been made into a TCR transgenic mouse. Unlike the BDC2.5 transgenic mice, BDC6.9 transgenic mice develop early diabetes by the 3rd and 4th generation backcross to the NOD background 178. The advantage to this clone is the ability to manipulate the presence and absence of self-Ag, as the stimulatory Ag is found in pancreas from NOD and SWR mice, but not other common strains of mice.

 

Daniel and Wegmann isolated a series of T cell clones from islet infiltrates and found that the majority reacted with insulin and the great majority of insulin-reactive (>90%) CD4 clones reacted with the insulin peptide B:9-23 (B chain, amino acids 9-23). A transgenic T cell receptor mouse has been established with the 12-4.1 T cell receptor anti-B:9-23 clone. This T cell recognizes insulin peptide sequences 13-23, using the common alpha chain motif (Valpha 13.3 (TRAV5D-4*04) with Jalpha 53)401. As a retrogenic the T cell receptor of the 12-4.4 clone is also able to induce insulin autoantibodies and diabetes 107, 397.  The 12-4.1 and 12-4.4 alpha chains have the same TRAV5D-4*04 T cell receptor segment, related J alpha segments, but different alpha chain N region and different Vbeta segments.  The ability of multiple T cell receptors with the related alpha chains that target the insulin B:9-23 peptide to induce insulin autoantibodies and diabetes has led to the hypothesis that such targeting is primarily driven by the shared alpha chain germline sequences 32.

 

Both Class II-restricted CD4+ and class-I restricted CD8+ T cell clones isolated in the laboratory of  Santamaria have been developed into TCR transgenic mice with either beta chains alone or in combination with alpha chains. A majority of CD8+ T cell clones isolated from the pancreas of pre-diabetic NOD mice show the same TCR Va17/Ja42 with N region sequence M-R-D/E, which has been used to generate 8.3 TCRab transgenic mice. The Vb8.1 CD8+ TCR transgenic mice, transgenic for the TCR beta chain only, and 8.3 CD8+ transgenic mice show an accelerated incidence of disease with a reduced period of benign insulitis 386. In the 8.1 beta-chain only transgenic mice, the islet-reactive T cells were found to have the same alpha chain as the original clone, suggesting a selection of this clone in diabetogenesis 386. The target antigen of the 8.3 clone is the molecule IGRP (islet-specific glucose-6-phosphatase catalytic subunit-related protein) and an excellent tetramer reagent (using a mimotope of the native peptide NRP-V7) allows staining of reactive T cells from peripheral blood of NOD mice, with numbers of T cells correlating with disease progression 208. Islet-reactive CD8+ T cells increase their avidity binding to the 8.3-reactive peptide presented in Kd as shown through tetramer analysis 209.  Kay and coworkers have recently found that the 8.3 transgenic induction of diabetes is dependent upon an immune response to insulin by blocking such a response with a proinsulin transgene5.

 

The NY4.1 transgenic mouse is from a CD4 clone. This TCR transgenic mouse has the same disease incidence on RAG2 KO or normal NOD background, confirming the ability of some CD4+ cells to induce diabetes in the absence of CD8+ T cells 386. However, breeding the Vb8.3 CD8+ TCRabtransgenic mice onto the RAG2 KO background results in greatly reduced disease onset and demonstrates a role for CD4 cells in the efficient recruitment of CD8+ T cells to islets 387. The NY4.1 as a retrogenic induces diabetes264. The transition from benign insulitis to overt diabetes appears to be a loss in the ability to regulate autoreactive cells. Epitope spreading and affinity maturation have been shown to be important in this process.

 

Previous reports demonstrated that a CD8+ T cell clone (G9C8) isolated from the pancreas of RIP-B7.1 transgenic mice did not require CD4 cells for disease transfer 389. In a more recent study a TCR transgenic mouse (NOD.AI4) was generated from a CD8 clone which was isolated from an unmanipulated NOD pancreas. The transgenic T cells were capable of inducing diabetes free of CD4 help or transgenic costimulation, supporting the idea that high-affinity CD8 T cells are independently capable of beta cell destruction 388. Of interest the clone apparently requires both Kd and Db to be present for cytotoxicity 84. However, another CD8+ islet-reactive TCR transgenic, the 9.3 transgenic, showed accelerated diabetes compared to normal NOD mice but no diabetes on the NOD.scid background 402. The unusual finding in this transgenic system was the necessity for two dissimilar receptors on the cell surface for disease incidence as identified by co-staining with antibodies specific for the transgene and a mix of other Vbeta-specific Antibodies. The authors conclude that the lowered expression of the pancreatic-specific receptor due to coexpression of other TCRs allowed escape of the transgenic receptors from negative selection. Adoptive transfer studies showed these cells did require CD4 help for diabetes induction 402.

 

            Vignali and coworkers have introduced a powerful new technique for the study of T cell receptor expression that they term “retrogenics” in distinction to transgenics. The use retroviral- mediated stem cell gene transfer of T cell receptor sequences and bone marrow transplantation of the transduced bone marrow. For the production of retrogenic mice a retrovirus is used to introduce genes coding for T Cell receptor chains into bone marrow cells that are then transplanted into immunodeficient mice (usually SCID or rag-/-).  Within weeks one can observe the development of insulin autoantibodies, insulitis and diabetes.  The figure (Nakayama) below illustrates the general methodology for production of transgenic mice where one can introduce a series of Valpha chains of anti-islet T cell receptors  that differ in different segments or complete alpha a beta chain T cell receptors. If the donor marrow is from a Calpha knockout mouse, the only alpha chain produced in the retrogenic mouse will be the introduced chain.  If the donor mouse is a SCID and complete alpha and beta TCR chains are introduced, only a single T cell receptor will be produced.

 

Vignali and coworkers studied two T cell receptor positive controls and showed that BDC2.5 and 4.1 retrogenic bone marrow induced diabetes in NOD-scid mice. In contrast T cell receptors for GAD206-220 and GAD524-538, though reactive cells were present in vivo, failed to influence the development of diabetes of NOD mice 396.  A subset of anti-GAD TCR retrogenic develop encephalitis (no insulitis).  More generally the retrogenic technique provides a powerful tool to rapidly study many T cell receptors in vivo264 .  Studies of multiple T cell receptors targeting different islet autoantigens indicate that only a subset lead to insulitis and less produce diabetes.  T cell receptors targeting the insulin B:9-23 peptide can cause disease, and a set of T cell receptors are more diabetogenic that react with currently unknown islet autoantigens.  It is likely that production of retrogenics will accelerate the study of T cell receptors and importantly segments of T cell receptors to determine targeting and pathogenicity.   For instance DiLorenzo and coworkers have utilized retrogenics to produce large numbers of CD8 T cells with specific T cell receptors to study in vitro393. A disadvantage of the retrogenic technique is that the introduced T cell receptors are naturally not heritable, and for each experiment new mice need to be created.

 

Transgenic Expression NeoSelf-Antigens in Pancreas

            A limitation for many of the studies using TCRs from pancreatic-reactive clones is the lack of knowledge of the antigen. This is a limitation that is rapidly being addressed, now with anti-insulin TCR transgenics (BDC12-4.1 TCR), IGRP (islet-specific glucose-6-phosphatase catalytic subunit-related protein) transgenics (8.3 TCR transgenics) 207, AI4 transgenics (?DMK target autoantigen) and BDC 2.5 and BDC10.1 transgenic (chromogranin) 244. Nevertheless, studies with defined antigens introduced into the pancreas by transgenesis on the RIP (rat insulin promoter) promoter, combined with T cell receptor transgenics have provided an elegant system for evaluating tolerance to islet expressed molecules. A list of models is present in Table 3.5. A important caveat, not recognized in the very initial studies of Rat Insulin Promoter driven Neo-Antigen expression is the expression of the transgene neoSelf-Antigen in lymphoid tissues such as the thymus. Thus autoimmunity can be influenced not only by antigen in the target tissue but dramatically by central tolerance mechanisms. The initial studies by Miller and coworkers with RIP induced expression of a “foreign” class I molecule, elegantly demonstrate the importance of minute expression within the thymus of neo-Self-Antigen, with tolerance dependent upon thymic expression of the antigen and not expression within islets 403.Transgenic expression of viral LCMV under the RIP promoter is associated with ignorance of the T cells to the LCMV antigen unless prompted by LCMV systemic infection which leads to diabetes 404, 405. It is hypothesized that this ignorance is due to poor cross-presentation of antigens such as gp33 by CD4 T cells 406. The diabetes in this model always requires CD8+ T cells whereas CD4+ T cells are only required in some lines 407-409. When the RIP-LCMV is crossed with a TCR transgenic mouse specific for LCMV, the animal still requires systemic LCMV infection for diabetes induction, although in this model the disease onset is accelerated compared to the non TCR transgenic model 405. In the LCMV transgenic model, the CD8+ T cells do not require CD4+ help as suggested by disease incidence with LCMV infection in class II-deficient 408 and CD40L-deficient animals  (120). As with other systems, crossing the RIP-LCMV with RIP-B7.1 transgenic mice, which provides costimulation directly on islet cells, results in spontaneous diabetes 410, 411. Recent work with a defined LCMV protein (NP) crossed with a RIP-IL4 transgenic mouse showed protection from diabetes following LCMV-infection 412. The IL4 was shown to inhibit the acquisition of cytolytic activity yet promote expansion and survival of CD8+ T cells through an increased expression of B7.2 on DCs 412.

 

Another neo self-Antigen model targets expression of flu hemagglutinin (HA) antigen to pancreatic beta cells using the RIP promoter. In one set of models HA expressed with Igk promoter results in expression in thymic medullary and cortical epithelial cells (as wells a B lymphocytes)

and thymuses induce CD25+ regulatory T cells 413. Unlike the LCMV protein, the Ins-HA mice appear tolerant to the HA Antigen following flu immunization if administered in adulthood 410, 414. A CD8+ TCR transgenic specific for HA (CL4) can adoptively transfer diabetes in Ins-HA recipients if the transgenic T cells are pre-activated in vitro 415. In addition, CL4xIns-HA double transgenic mice are not tolerant and get spontaneous diabetes by 8 days of age with few T cells in the islet infiltrate 414. Further work on the CL4 TCR showed this TCR possessed an unusually high affinity to HA/Kd.

 

 

 

 

Table 3.5 “Neo-Self Antigen Models to Type 1 Diabetes

This high affinity is evident in the activation of the TCR in the absence of costimulation or with soluble monomeric complexes 415. Like this CD8+ TCR transgenic mouse, tolerance was also not established to the HA Antigen in mice doubly transgenic for the Ins-HA transgenic and a CD4+ anti-HA TCR transgenic 416. Other HA-specific CD4+ TCR transgenic mice crossed with Ins-HA developed varying degrees of diabetes depending on the background strain 416, 417. Recently, the Ins-HA model has been used in studies of peripheral tolerance of CD8+ T cells in various mouse studies. Defects in peripheral tolerance in NOD mice compared to BALB/c and B10.D2 were confirmed by showing higher avidity binding of NOD T cells to Kd-HA tetramers 205.

 

Similar to the Ins-HA model, RIP-OVA transgenic mice can become diabetic when transferred with pre-activated anti-OVA CD8+ T cells 418. In addition, early diabetes is observed in OT-1 (TCR transgenic anti-OVA)xRIP-OVA double transgenic mice, suggesting a lack of tolerance and ignorance in this model 419. Another model places the class I Kb molecule under control of the RIP-promoter 403. Crossing the RIP-Kb mice with a CD8+ TCR anti-Kb transgenic mouse shows tolerance to this pancreatic islet, which is associated with both thymic deletion of high-affinity clones by residual thymic expression and functional unresponsiveness of remaining clones. This tolerance can be broken, resulting in diabetes induction, with RIP expression of IL2, a cytokine which has been shown to reverse T cell anergy 420.

 

            Transgenic expression of the exogenous hen egg lysozyme (HEL) protein by the RIP promoter, together with an anti-HEL TCR transgenic, shows thymic deletion and peripheral anergy of transgenic cells on the C57BL/6 background. However when bred onto the NOD background, maintaining the necessary H-2k MHC, the HEL model showed defects in both thymic deletion and T cell anergy induction leading to overt diabetes in the NOD RIP-HEL/TCR double-transgenic mice. This model clearly demonstrates the influence of NOD non-MHC genes on tolerance processes 421.

 

            As mentioned above, transgenic mice expressing the costimulatory molecule B7.1 on pancreatic islet cells (RIP-B7.1) have been useful in studying “spontaneous” diabetes by crossing this mouse with models which otherwise do not show disease incidence. This mouse on the C57BL/6 background does not get diabetes but can become diabetic when crossed with RIP-LCMV, RIP-TNFa, or RIP-IE or with mice expressing human HLA molecules associated with type 1 diabetes. The usefulness of the RIP-B7.1 model is to address the sensitivity of disease induction allowing the systems to be stressed to determine if diabetes can occur. RIP-B7.1 transgenic expression on the NOD background results in diabetes incidence as early as the first backcross although IAg7 homozygosity is required 415. The largest effect of B7.1 expression is on the CD8 cells, thereby negating the necessity of CD4+ help although the presence of CD4+ cells accelerates the disease progression by aiding in migration of cells to the pancreas. Unlike spontaneous NOD diabetes, B cells are not required for disease in the RIP-B7.1 model 422.

 

Immunization/Immunoregulatory Induced Models of Diabetes

 

 

            For many experimental autoimmune disorders it is possible to induce pathology by immunizing with the presumed target autoantigen, creating disorders such as Experimental Autoimmune Encephalitis (EAE) 122. Until recently it has not been possible to induce immune mediated diabetes in mice or rats with specific islet autoantigens. Immunization with a heat shock protein was associated with transient hyperglycemia 423 and this molecule, despite its ubiquitous expression is reported to be both an autoantigen and activator of the innate immune system 245.

 

There is actually an “old” literature where insulin immunization of cows and rabbits induced “insulitis” and even diabetes in rabbits 424-429but with the “difficulty” of the bovine model, and apparently problems in reproducibly inducing diabetes in the rabbit model, (as available insulin became purer), this area of investigation was not pursued. It was certainly recognized that most patients treated with insulin (again becoming less with improved insulin purity) produced insulin antibodies and immunoreactivity to insulin was studied in mouse models 430 and man 431. It is somewhat surprising that it was not possible to induce diabetes with proinsulin/insulin/or insulin peptides. Recent studies indicate that insulin and its peptides can induce anti-islet autoimmunity (insulin autoantibodies and insulitis), but in normal mouse strains (e.g., BALB/c, C57BL/6) this is not sufficient to engender diabetes. In mice carrying a transgene inducing B7.1 (costimulatory molecule) expression on islets, diabetes is routinely induced following insulin/ insulin peptide B:9-23 immunization given the appropriate MHC alleles 232, 432-435.

 

 

Figure 7. Induction of insulin autoantibodies following subcutaneous injection of insulin peptide B:9-23 in incomplete Freund’s adjuvant.

 

Mice with I-Ad (e.g., BALB/c mice) or I-Ag7 (e.g. NOD mice) respond to subcutaneous immunization with insulin peptide B:9-23 with the rapid production of insulin autoantibodies (Figure 7) 432-434. NOD mice respond in a similar fashion, but because they already produce insulin autoantibodies, one observes increased levels rather than de novo production of autoantibodies. In NOD mice the expression of insulin autoantibodies persist for months, rather than being transient following immunization with B:9-23 peptide. These are actual autoantibodies in that they can be blocked with “mouse” insulin, and the antibodies react with intact insulin and cannot be absorbed with the immunizing B:9-23 peptide. Of note the response is MHC restricted as shown by analysis of congenic strains and no other peptide of proinsulin to date has been found to induce the autoantibodies.

 

When NOD mice are immunized with the insulin peptide B:9-23 they also produce antibodies reacting with the peptide itself, that depending on the chronicity of administration can induce anaphylaxis 436, 437. Though insulin autoantibodies are induced by insulin peptide B:9-23 immunization of BALB/c mice, insulitis was not observed. To produce insulitis, immunization of BALB/c mice was combined with two weeks of administration of subcutaneous poly-IC. Poly-IC is a toll 3 receptor activator and mimic of double stranded RNA. It induces circulating levels of interferon alpha, and acts through induction of interferon alpha to induce diabetes438.   Despite the development of both insulin autoantibodies and insulitis, the BALB/c mice do not progress to diabetes. In mice expressing B7.1 in islet beta cells, diabetes can be induced with either poly-IC or insulin B:9-23 peptide alone, as well as with both together 433. Thus tolerance to insulin can be readily broken, and there is a dramatic restriction in terms of the peptides able to induce disease. We suspect that normal BALB/c mice are already sensitized to the insulin B:9-23 peptide prior to immunization with this peptide given the rapidity of the induction of insulin autoantibodies reacting with a conformational epitope of insulin. For the C57Bl6 mouse that does not respond to insulin peptide B:9-23 (I-Ab) disease can be induced in B7.1 islet transgenic mice by immunization with intact insulin, presumably by reactivity to a different epitope of insulin 435.  Induction of cytokines can also accelerate the development of diabetes in NOD mice depending upon the stage at which for instance NOD mice with established insulitis when infected with rotavirus439.

 

            Co-stimulation

 

            As mentioned previously, the expression of the B7.1 molecule on islets enhances the diabetogenicity of a series of stimuli that by themselves are insufficient to induce diabetes. This includes insulin peptide B:9-23 (BALB/c mice) 433, poly-IC (C57BL/6 mice) 440, LCMV-GP peptide 441, LCMV-GP gp33 and preproinsulin encoding plasmid 442, human DR4 transgene 443, human DQ8 transgene 443, transgenic islet expression of IL2 444, and transgene induced expression of TNF alpha on beta cells 445. It was hypothesized the B7-H1 would be a negative regulator of autoimmunity, but transgene induced expression of B7-H1 on islets also induced diabetes 446.

 

“Humanized” Mice

            Another group of animal models being developed in the past decade for the study of type 1 diabetes are the humanized mice. There are two general approaches:  1. Introduction of human genes into mice and 2. Transplantation of human stem cells into immunodeficient mice19.  Both approaches represent important technical advances20, 447, 448.

 

            The class II HLA molecule DQB1 chain with serine, alanine or valine at P57 of the b chain, similar to the NOD MHC class II haplotype, confers the greatest risk for type 1 diabetes in humans. The DQ8 (DQA1*0301 plus DQB1*0302 (alanine at P57)) haplotype has been expressed in transgenic mice 449. The DQ8 transgenic mice do not develop diabetes even when backcrossed to the NOD background. However, when these mice are crossed with the RIP-B7.1 transgenic mice, 80% of the mice develop insulitis and type 1 diabetes whereas control human DQ6 (diabetes-resistant haplotype)xRIP-B7.1 double-transgenic mice do not develop insulitis or diabetes. The DQ8xRIP-B7.1 transgenic mice have been used to map approximately 10 different T cell epitopes in human GAD65, one of which is also a dominant T cell epitope in NOD mice restricted to IAg7 450, 451. Transgenic expression of the human disease-associated DR4 allele in mice showed surprisingly that this allele, in combination with the highly susceptible DQ8 allele, actually protected mice from disease. Whereas 25% of DR4/RIP-B7 mice developed diabetes compared to >80% of DQ8/RIP-B7 mice, the triple transgenic DR4/DQ8/RIP-B7 had only a 23% disease incidence, showing a dominant protective effect for the DR4 allele  (137). The diabetes incidence afforded by the immunostimulatory RIP-B7.1 transgene enables studies of human autoimmunity genes in mice with the important caveat that the pattern of disease can be very unusual as found by Lipes and coworkers with pancreatitis depending on the strain in the absence of murine class II genes 452. RIP-B7/DRB1*0404 HLA transgenic mice spontaneously develop T cells reacting with GAD65 and Glial fibrillary acid protein (GFAP) and approximately 20% develop diabetes at a mean age of 40 weeks453. A human T cell receptor transgenic mouse (anti GAD65/67 (555-567) develops insulitis on a human DR4 rag-/- background268.

            The introduction of human DQ8 onto the NOD background has resulted in a series of autoimmune disorders. In particular a disorder resembling dermatitis herpetiformis is induced followed immunization with gluten 454, a disorder resembling polychondritis following immunization with type II collagen 455.

            Marron and coworkers demonstrated that transgenic expression of the common human class I allele, HLA-A2.1, accelerated the development of diabetes in NOD mice. This effect was not simply a result of expression of any human class I allele, as HLA-B27 (associated with ankylosing spondylitis in man) if anything decreased development of diabetes 456. The A2.1 mouse islets have been used to study a model of human lymphocytes in NOD-Rag1 (null)Prf1 (null) mice rejecting these islets. Transplanting both into the spleen enhanced graft and lymphocyte interaction. When lymphocytes came from A2.1 positive donors the grafts survived, while from HA-A2 negative donors, the grafts were rejected 457.  Humanized HLA-A2.1 mice (further refined to eliminate murine class I antigens) have been utilized to identify peptides recognized by CD8 T cells of potential relevance to human type 1 diabetes447, 458, 459.

 

Human Stem Cell Transplantation

            It is likely that many additional human molecules will be introduced into murine strains to “humanize” the immune system.  An approach to utilize transplantation/transfer of human cells directly into mice allows the rapid introduction of multiple genes and cell types20.  These studies have been greatly enhanced with the development of common gamma chain knockout mice on the NOD-scid background460. This combines severe adaptive immunodeficiency with abnormalities of NK T cells and allows engraftment of both peripheral blood cells and stem cells and the study of alloreactivity with transplant rejection19.  This model has been combined with the Akita mutation resulting in diabetes and when human hematopoietic stem cells are engrafted approximately ½ of mice with transplanted islets reject their human islet allograft 461.

 

Conclusion

            The multiple animal models of type 1A (immune mediated) diabetes amply demonstrate that there are many pathways that can result in immune mediated (autoimmune) destruction of islet beta cells, and that the spontaneous animal models each represent a different pathway. One can generalize with the following observations:

 

  1. T cell destruction of islet beta cells can be entrained by the targeting of single islet molecular determinants by T cells with a single T cell receptor (and obviously also with multiple receptors). In general however, both CD4 and CD8 T cells collaborate in causing beta cell destruction. It is likely that each T cell receptor (e.g., clone or transgenic) will follow its own rules, depending on the avidity and other qualities of the receptor), in terms of whether ignorance, tolerance or beta cell destruction results.

 

  1. There is often T cell receptor biased chain utilization of the T cells targeting a specific peptide autoantigen.

 

  1. In the spontaneous animal models multiple islet antigens are the targets of autoimmunity.

 

  1. It is likely in some common models, such as the NOD mouse, a single determinant may be essential for disease, such as the insulin sequence B:9-23.

 

  1. The finding that a given antigen is the target of autoreactive T lymphocytes, and even that T cells targeting that antigen can transfer disease, does not mean that the targeted molecule is important for pathogenesis of the spontaneous disorder, and the best current test for importance of a target epitope, is gene knockout or mutation. If disease occurs at the same rate in the absence of the molecule, it cannot be essential, though may contribute  to the disorder.

 

  1. Multiple genes synergistically contribute to autoimmunity and the MHC is often essential in determining the organ targeted.

 

  1. Both pathogenic T cells and regulatory T cells (and other regulatory cells) modulate the development of disease.

 

The tremendous increase in knowledge relative to the animal models will hopefully translate into informed trials for the prevention of type 1A diabetes in man and long-term success of islet transplantation. There is an important opportunity to attempt to develop assays and paradigms in the animal models that are directly applicable to man. In particular in man one must sample the peripheral blood for disease prediction, and every effort should be made in the “simpler” animal models to develop assays of blood that can be applied to man.

 

 

 

 

 

 

 

 

 

 

Reference List

 

    (1)   Stadinski BD, Zhang L, Crawford F, Marrack P, Eisenbarth GS, Kappler JW. Diabetogenic T cells recognize insulin bound to IAg7 in an unexpected, weakly binding register. Proc Natl Acad Sci U S A 2010 Jun 15;107(24):10978-83.PM:20534455

    (2)   Stadinski BD, Delong T, Reisdorph N, Reisdorph R, Powell RL, Armstrong M, et al. Chromogranin A is an autoantigen in type 1 diabetes. Nat Immunol 2010 Mar;11(3):225-31.PM:20139986

    (3)   Lang J, Bellgrau D. Animal models of type 1 diabetes: genetics and immunological function. Adv Exp Med Biol 2004;552:91-116.http://www.ncbi.nlm.nih.gov/pubmed/15622960?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

    (4)   Nakayama M, Abiru N, Moriyama H, Babaya N, Liu E, Miao D, et al. Prime role for an insulin epitope in the development of type 1 diabetes in NOD mice. Nature 2005 May 12;435(7039):220-3.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=pubmed&cmd=Retrieve&dopt=AbstractPlus&list_uids=15889095

    (5)   Krishnamurthy B, Mariana L, Gellert SA, Colman PG, Harrison LC, Lew AM, et al. Autoimmunity to Both Proinsulin and IGRP Is Required for Diabetes in Nonobese Diabetic 8.3 TCR Transgenic Mice. J Immunol 2008 Apr 1;180(7):4458-64.http://www.ncbi.nlm.nih.gov/pubmed/18354167?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

    (6)   Su MA, Giang K, Zumer K, Jiang H, Oven I, Rinn JL, et al. Mechanisms of an autoimmunity syndrome in mice caused by a dominant mutation in Aire. J Clin Invest 2008 May;118(5):1712-26.http://www.ncbi.nlm.nih.gov/pubmed/18414681?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

    (7)   Chen Z, Herman AE, Matos M, Mathis D, Benoist C. Where CD4+CD25+ T reg cells impinge on autoimmune diabetes. J Exp Med 2005 Nov 21;202(10):1387-97.http://www.ncbi.nlm.nih.gov/pubmed/16301745?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

    (8)   Mordes JP, Bortell R, Blankenhorn EP, Rossini AA, Greiner DL. Rat models of type 1 diabetes: genetics, environment, and autoimmunity. ILAR J 2004;45(3):278-91.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15229375

    (9)   Leiter EH. Mice with targeted gene disruptions or gene insertions for diabetes research: problems, pitfalls, and potential solutions. diabetol 2002 Mar;45(3):296-308.http://www.ncbi.nlm.nih.gov/pubmed/11914735?ordinalpos=7&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (10)   Roep BO, Atkinson M, von Herrath M. Satisfaction (not) guaranteed: re-evaluating the use of animal models of type 1 diabetes. Nat Rev Immunol 2004 Dec;4(12):989-97.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15573133

  (11)   Anderson MS, Bluestone JA. The NOD Mouse: A Model of Immune Dysregulation. Annu Rev Immunol 2004 Nov 29.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15568997

  (12)   Devendra D, Liu E, Eisenbarth GS. Type 1 diabetes: recent developments. Brit Med J 2004;328(7442):750-4.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=pubmed&cmd=Retrieve&dopt=AbstractPlus&list_uids=15044291&query_hl=1&itool=pubmed_docsum

  (13)   Eisenbarth GS, Gottlieb PA. Autoimmune polyendocrine syndromes. N Engl J Med 2004 May 13;350(20):2068-79.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=pubmed&cmd=Retrieve&dopt=AbstractPlus&list_uids=15141045&query_hl=3&itool=pubmed_docsum

  (14)   Homann D, von Herrath M. Regulatory T cells and type 1 diabetes. Clin Immunol 2004 Sep;112(3):202-9.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15308110

  (15)   Mathis D, Benoist C. Back to central tolerance. Immunity 2004 May;20(5):509-16.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15142520

  (16)   Panagiotopoulos C, Trudeau JD, Tan R. T-cell epitopes in type 1 diabetes. Curr Diab Rep 2004 Apr;4(2):87-94.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15035967

  (17)   Polychronakos C. Animal models of spontaneous autoimmune diabetes: notes on their relevance to the human disease. Curr Diab Rep 2004 Apr;4(2):151-4.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15035976

  (18)   Ramsdell F. Foxp3 and natural regulatory T cells: key to a cell lineage? Immunity 2003 Aug;19(2):165-8.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=12932350

  (19)   King M, Pearson T, Shultz LD, Leif J, Bottino R, Trucco M, et al. Development of new-generation HU-PBMC-NOD/SCID mice to study human islet alloreactivity. Ann N Y Acad Sci 2007 Apr;1103:90-3. Epub@2007 Mar 21.:90-3.http://www.ncbi.nlm.nih.gov/pubmed/17376822?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (20)   Pearson T, Greiner DL, Shultz LD. Creation of "humanized" mice to study human immunity. Curr Protoc Immunol 2008 May;Chapter 15:Unit 15.21.:Unit.http://www.ncbi.nlm.nih.gov/pubmed/18491294?ordinalpos=8&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (21)   Enee E, Martinuzzi E, Blancou P, Bach JM, Mallone R, van Endert P. Equivalent specificity of peripheral blood and islet-infiltrating CD8+ T lymphocytes in spontaneously diabetic HLA-A2 transgenic NOD mice. J Immunol 2008 Apr 15;180(8):5430-8.http://www.ncbi.nlm.nih.gov/pubmed/18390725?ordinalpos=19&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (22)   Kasprowicz DJ, Smallwood PS, Tyznik AJ, Ziegler SF. Scurfin (FoxP3) controls T-dependent immune responses in vivo through regulation of CD4+ T cell effector function. J Immunol 2003 Aug 1;171(3):1216-23.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=12874208

  (23)   Anderson MS, Venanzi ES, Klein L, Chen Z, Berzins SP, Turley SJ, et al. Projection of an immunological self shadow within the thymus by the aire protein. Science 2002 Nov 15;298(5597):1395-401.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=12376594

  (24)   Liston A, Lesage S, Wilson J, Peltonen L, Goodnow CC. Aire regulates negative selection of organ-specific T cells. Nat Immunol 2003 Apr;4(4):350-4.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=12612579

  (25)   Yang Y, Santamaria P. Lessons on autoimmune diabetes from animal models. Clin Sci (Lond) 2006 Jun;110(6):627-39.http://www.ncbi.nlm.nih.gov/pubmed/16689681?ordinalpos=6&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (26)   Roep BO. Are insights gained from NOD mice sufficient to guide clinical translation? Another inconvenient truth. Ann N Y Acad Sci 2007 Apr;1103:1-10. Epub@2007 Mar 21.:1-10.http://www.ncbi.nlm.nih.gov/pubmed/17376838?ordinalpos=31&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (27)   Shoda LK, Young DL, Ramanujan S, Whiting CC, Atkinson MA, Bluestone JA, et al. A comprehensive review of interventions in the NOD mouse and implications for translation. Immunity 2005 Aug;23(2):115-26.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=16111631

  (28)   Herold KC, Hagopian W, Auger JA, Poumian-Ruiz E, Taylor L, Donaldson D, et al. Anti-CD3 monoclonal antibody in new-onset type 1 diabetes mellitus. N Engl J Med 2002 May 30;346(22):1692-8.PM:12037148

  (29)   Eisenbarth SC, Flavell RA. Innate instruction of adaptive immunity revisited: the inflammasome. EMBO Mol Med 2009 May;1(2):92-8.PM:20049709

  (30)   Moriyama H, Abiru N, Paronen J, Sikora K, Liu E, Miao D, et al. Evidence for a primary islet autoantigen (preproinsulin 1) for insulitis and diabetes in the nonobese diabetic mouse. Proc Natl Acad Sci U S A 2003 Sep 2;100(18):10376-81.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=pubmed&cmd=Retrieve&dopt=Abstract&list_uids=12925730

  (31)   Perone MJ, Bertera S, Shufesky WJ, Divito SJ, Montecalvo A, Mathers AR, et al. Suppression of autoimmune diabetes by soluble galectin-1. J Immunol 2009 Mar 1;182(5):2641-53.PM:19234158

  (32)   Homann D, Eisenbarth GS. An immunologic homunculus for type 1 diabetes. J Clin Invest 2006 May;116(5):1212-5.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=pubmed&cmd=Retrieve&dopt=AbstractPlus&list_uids=16670763

  (33)   Santamaria P. The long and winding road to understanding and conquering type 1 diabetes. Immunity 2010 Apr 23;32(4):437-45.PM:20412754

  (34)   Eisenbarth GS. Banting Lecture 2009: An Unfinished Journey: Molecular Pathogenesis to Prevention of Type 1A Diabetes. diab 2010 Apr;59(4):759-74.PM:20350969

  (35)   Singh B, Delovitch TL. Immune mechanisms that regulate susceptibility to autoimmune type I diabetes. Clin Rev Allergy Immunol 2000 Dec;19(3):247-64.http://www.ncbi.nlm.nih.gov/pubmed/11138408?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (36)   Zhang L, Nakayama M, Eisenbarth GS. Insulin as an autoantigen in NOD/human diabetes. Curr Opin Immunol 2008 Jan 4;.http://www.ncbi.nlm.nih.gov/pubmed/18178393?ordinalpos=28&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (37)   Driver JP, Serreze DV, Chen YG. Mouse models for the study of autoimmune type 1 diabetes: a NOD to similarities and differences to human disease. Semin Immunopathol 2011 Jan;33(1):67-87.PM:20424843

  (38)   Chaparro RJ, DiLorenzo TP. An update on the use of NOD mice to study autoimmune (Type 1) diabetes. Expert Rev Clin Immunol 2010 Nov;6(6):939-55.PM:20979558

  (39)   Giarratana N, Penna G, Adorini L. Animal models of spontaneous autoimmune disease: type 1 diabetes in the nonobese diabetic mouse. Methods Mol Biol 2007;380:285-311.:285-311.http://www.ncbi.nlm.nih.gov/pubmed/17876100?ordinalpos=17&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (40)   Ablamunits V, Sherry NA, Kushner JA, Herold KC. Autoimmunity and beta cell regeneration in mouse and human type 1 diabetes: the peace is not enough. Ann N Y Acad Sci 2007 Apr;1103:19-32. Epub@2007 Mar 21.:19-32.http://www.ncbi.nlm.nih.gov/pubmed/17376841?ordinalpos=30&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (41)   Di Lorenzo 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 Apr;148(1):1-16.http://www.ncbi.nlm.nih.gov/pubmed/17349009?ordinalpos=34&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (42)   Stadinski B, Kappler J, Eisenbarth GS. Molecular targeting of islet autoantigens. Immunity 2010 Apr 23;32(4):446-56.PM:20412755

  (43)   Fierabracci A. Peptide immunotherapies in Type 1 diabetes: lessons from animal models. Curr Med Chem 2011;18(4):577-86.PM:21143110

  (44)   Wicker LS, Clark J, Fraser HI, Garner VE, Gonzalez-Munoz A, Healy B, et al. Type 1 diabetes genes and pathways shared by humans and NOD mice. J Autoimmun 2005;25 Suppl:29-33. Epub@2005 Oct 28.:29-33.http://www.ncbi.nlm.nih.gov/pubmed/16257508?ordinalpos=74&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (45)   Suri A, Unanue ER. The Murine Diabetogenic Class II Histocompatibility Molecule I-A(g7): Structural and Functional Properties and Specificity of Peptide Selection. Adv Immunol 2005;88:235-65.:235-65.http://www.ncbi.nlm.nih.gov/pubmed/16227092?ordinalpos=76&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (46)   Haskins K. Pathogenic T-cell clones in autoimmune diabetes: more lessons from the NOD mouse. Adv Immunol 2005;87:123-62.:123-62.http://www.ncbi.nlm.nih.gov/pubmed/16102573?ordinalpos=88&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (47)   Chatenoud L, Bach JF. Regulatory T cells in the control of autoimmune diabetes: the case of the NOD mouse. Int Rev Immunol 2005 May;24(3-4):247-67.http://www.ncbi.nlm.nih.gov/pubmed/16036377?ordinalpos=93&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (48)   Hussain S, Wagner M, Ly D, Delovitch TL. Role of regulatory invariant CD1d-restricted natural killer T-cells in protection against type 1 diabetes. Immunol Res 2005;31(3):177-88.http://www.ncbi.nlm.nih.gov/pubmed/15888910?ordinalpos=99&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (49)   Leiter EH. Nonobese diabetic mice and the genetics of diabetes susceptibility. Curr Diab Rep 2005 Apr;5(2):141-8.http://www.ncbi.nlm.nih.gov/pubmed/15794919?ordinalpos=101&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (50)   Anderson MS, Bluestone JA. The NOD mouse: a model of immune dysregulation. Annu Rev Immunol 2005;23:447-85.:447-85.http://www.ncbi.nlm.nih.gov/pubmed/15771578?ordinalpos=103&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (51)   Babad J, Geliebter A, DiLorenzo TP. T-cell autoantigens in the non-obese diabetic mouse model of autoimmune diabetes. Immunology 2010 Dec;131(4):459-65.PM:21039471

  (52)   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.http://www.ncbi.nlm.nih.gov/pubmed/6995140?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (53)   Kanazawa Y, Komeda K, Sato S, Mori S, Akanuma K, Takaku F. Non-obese-diabetic mice: immune mechanisms of pancreatic B-cell destruction. diabetol 1984;27:113-5.http://www.ncbi.nlm.nih.gov/pubmed/6383913?ordinalpos=15&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (54)   Wicker LS, Miller BJ, Coker LZ, McNally SE, Scott S, Mullen Y, et al. Genetic control of diabetes and insulitis in the nonobese diabetic (NOD) mouse. J Exp Med 1987;165:1639-54.http://www.ncbi.nlm.nih.gov/pubmed/3585250?ordinalpos=4&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (55)   Maruyama T, Takei I, Yanagawa T, Takahashi T, Asaba Y, Kataoka K, et al. Insulin autoantibodies in non-obese diabetic (NOD) mice and streptozotocin induced diabetic mice. Diabetes Res 1988;7(2):93-6.http://www.ncbi.nlm.nih.gov/pubmed/3293880?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (56)   Ziegler AG, Vardi P, Ricker AT, Hattori M, Soeldner JS, Eisenbarth GS. Radioassay determination of insulin autoantibodies in NOD mice. Correlation with increased risk of progression to overt diabetes. diab 1989;38(3):358-63.http://www.ncbi.nlm.nih.gov/pubmed/2917700?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (57)   Gottlieb PA, Eisenbarth GS. Mouse and man: multiple genes and multiple autoantigens in the aetiology of type I DM and related autoimmune disorders. J Autoimmun 1996;9:277-81.http://www.ncbi.nlm.nih.gov/pubmed/8738974?ordinalpos=5&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (58)   Ott PA, Anderson MR, Tary-Lehmann M, Lehmann PV. CD4+CD25+ regulatory T cells control the progression from periinsulitis to destructive insulitis in murine autoimmune diabetes. Cell Immunol 2005 May;235(1):1-11.http://www.ncbi.nlm.nih.gov/pubmed/16122720?ordinalpos=4&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (59)   Todd JA, Aitman TJ, Cornall RJ, Ghosh S, Hall JR, Hearne CM, et al. Genetic analysis of autoimmune type I diabetes mellitus in mice. Nature 1991;351(6327):542-7.http://www.ncbi.nlm.nih.gov/pubmed/1675432?ordinalpos=17&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (60)   Oldstone MBA. Prevention of type 1 diabetes in nonobese diabetic mice by virus infection. Science 1988;239:500-2.http://www.ncbi.nlm.nih.gov/pubmed/3277269?ordinalpos=14&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (61)   Wilberz S, Partke HJ, Dagnaes-Hansen F, Herberg L. Persistent MHV (mouse hepatitis virus) infection reduces the incidence of diabetes mellitus in non-obese diabetic mice. diabetol 1991;34(1):2-5.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=1647335

  (62)   Atkinson MA, Leiter EH. The NOD mouse model of type 1 diabetes: as good as it gets? Nat Med 1999 Jun;5(6):601-4.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=10371488

  (63)   Rainbow DB, Esposito L, Howlett SK, Hunter KM, Todd JA, Peterson LB, et al. Commonality in the genetic control of Type 1 diabetes in humans and NOD mice: variants of genes in the IL-2 pathway are associated with autoimmune diabetes in both species. Biochem Soc Trans 2008 Jun 1;36(3):312-5.http://www.ncbi.nlm.nih.gov/pubmed/18481948?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (64)   Hattori M, Buse JB, Jackson RA, Glimcher L, Dorf ME, Minami M, et al. The NOD mouse: recessive diabetogenic gene within the major histocompatibility complex. Science 1986;231(4739):733-5.http://www.ncbi.nlm.nih.gov/pubmed/3003909?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (65)   Todd JA, Bell JI, McDevitt HO. HLA-DQB gene contributes to susceptibility and resistance to insulin-dependent diabetes mellitus. Nature 1987;329(6140):599-604.http://www.ncbi.nlm.nih.gov/pubmed/3309680?ordinalpos=9&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (66)   Wicker LS, Todd JA, Peterson LB. Genetic control of autoimmune diabetes in the NOD mouse. Ann Rev Immunol 1995;13:179-200.http://www.ncbi.nlm.nih.gov/pubmed/7612220?ordinalpos=18&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (67)   Todd JA, Acha-Orbea H, Bell JI, Chao N, Fronek Z, Jacob CO, et al. A molecular basis for MHC class II associated autoimmunity. Science 1988;240(4855):1003-9.http://www.ncbi.nlm.nih.gov/pubmed/3368786?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (68)   Lund T, O'Reilly L, Hutchings P, Kanagawa O, Simpson E, Gravely R, et al. Prevention of insulin-dependent diabetes mellitus in non-obese diabetic mice by transgenes encoding modified I-A beta-chain or normal I-E alpha-chain. Nature 1990;345(6277):727-9.http://www.ncbi.nlm.nih.gov/pubmed/2163026?ordinalpos=12&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (69)   Inoue K, Ikegami H, Fujisawa T, Noso S, Nojima K, Babaya N, et al. Allelic variation in class I K gene as candidate for a second component of MHC-linked susceptibility to type 1 diabetes in non-obese diabetic mice. diabetol 2004 Apr;47(4):739-47.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15298352

  (70)   Fujisawa T, Ikegami H, Noso S, Yamaji K, Nojima K, Babaya N, et al. MHC-linked susceptibility to type 1 diabetes in the NOD mouse: further localization of Idd16 by subcongenic analysis. Ann N Y Acad Sci 2006 Oct;1079:118-21.:118-21.http://www.ncbi.nlm.nih.gov/pubmed/17130541?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (71)   Yamaji K, Ikegami H, Fujisawa T, Noso S, Nojima K, Babaya N, et al. Contribution of class III MHC to susceptibility to type 1 diabetes in the NOD mouse. Ann N Y Acad Sci 2006 Oct;1079:114-7.:114-7.http://www.ncbi.nlm.nih.gov/pubmed/17130540?ordinalpos=4&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (72)   Mohan JF, Levisetti MG, Calderon B, Herzog JW, Petzold SJ, Unanue ER. Unique autoreactive T cells recognize insulin peptides generated within the islets of Langerhans in autoimmune diabetes. Nat Immunol 2010 Apr;11(4):350-4.PM:20190756

  (73)   Ueda H, Howson JM, Esposito L, Heward J, Snook H, Chamberlain G, et al. Association of the T-cell regulatory gene CTLA4 with susceptibility to autoimmune disease. Nature 2003 Apr 30;423(6939):506-11.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=12724780

  (74)   Yamanouchi J, Rainbow D, Serra P, Howlett S, Hunter K, Garner VE, et al. Interleukin-2 gene variation impairs regulatory T cell function and causes autoimmunity. Nat Genet 2007 Feb 4;.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=pubmed&cmd=Retrieve&dopt=AbstractPlus&list_uids=17277778&query_hl=15&itool=pubmed_docsum

  (75)   Hamilton-Williams EE, Serreze DV, Charlton B, Johnson EA, Marron MP, Mullbacher A, et al. Transgenic rescue implicates beta2-microglobulin as a diabetes susceptibility gene in nonobese diabetic (NOD) mice. Proc Natl Acad Sci U S A 2001 Sep 25;98(20):11533-8.http://www.ncbi.nlm.nih.gov/pubmed/11572996?ordinalpos=12&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (76)   Todd JA, Wicker LS. Genetic protection from the inflammatory disease type 1 diabetes in humans and animal models. Immunity 2001 Sep;15(3):387-95.http://www.ncbi.nlm.nih.gov/pubmed/11567629?ordinalpos=10&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (77)   Robles DT, Eisenbarth GS, Dailey NJM, Peterson LB, Wicker LS. Insulin autoantibodies are associated with islet inflammation but not always related to diabetes progression in NOD congenic mice. diab 2002;52(3):882-6.http://www.ncbi.nlm.nih.gov/pubmed/12606534?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (78)   Encinas JA, Kuchroo VK. Mapping and identification of autoimmunity genes. Curr Opin Immunol 2000 Dec;12(6):691-7.http://www.ncbi.nlm.nih.gov/pubmed/11102774?ordinalpos=4&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (79)   McDuffie M. Genetics of autoimmune diabetes in animal models. Curr Opin Immunol 1998 Dec;10(6):704-9.http://www.ncbi.nlm.nih.gov/pubmed/9914220?ordinalpos=8&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (80)   Wakeland E, Morel L, Achey K, Yui M, Longmate J. Speed congenics: a classic technique in the fast lane (relatively speaking). Immunol Today 1997;18:472-7.http://www.ncbi.nlm.nih.gov/pubmed/9357138?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (81)   Brook FA, Evans EP, Lord CJ, Lyons PA, Rainbow DB, Howlett SK, et al. The derivation of highly germline-competent embryonic stem cells containing NOD-derived genome. diab 2003 Jan;52(1):205-8.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=12502514

  (82)   Kawabata Y, Ikegami H, Fujisawa T, Noso S, Asano K, Hiromine Y, et al. A second component of HLA-linked susceptibility to type 1 diabetes maps to class I region. Ann N Y Acad Sci 2006 Oct;1079:278-84.:278-84.http://www.ncbi.nlm.nih.gov/pubmed/17130566?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (83)   Ikegami H, Fujisawa T, Makino S, Ogihara T. Congenic mapping and candidate sequencing of susceptibility genes for Type 1 diabetes in the NOD mouse. Ann N Y Acad Sci 2003 Nov;1005:196-204.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=14679059

  (84)   Takaki T, Lieberman SM, Holl TM, Han B, Santamaria P, Serreze DV, et al. Requirement for both H-2Db and H-2Kd for the induction of diabetes by the promiscuous CD8+ T cell clonotype AI4. J Immunol 2004 Aug 15;173(4):2530-41.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15294969

  (85)   Acha-Orbea H, McDevitt HO. The first external domain of the nonobese diabetic mouse class II I-A chain is unique. Proc Natl Acad Sci USA 1987;84(8):2435-9.http://www.ncbi.nlm.nih.gov/pubmed/2882518?ordinalpos=15&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (86)   Nepom GT, Erlich H. MHC class-II molecules and autoimmunity. Ann Rev Immunol 1991;9:493-525.http://www.ncbi.nlm.nih.gov/pubmed/1910687?ordinalpos=53&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (87)   Miyazaki T, Uno M, Uehira M, Kikutani H, Kishimoto T, Kimoto M, et al. Direct evidence for the contribution of the unique I-ANOD to the development of insulitis in non-obese diabetic mice. Nature 1990 Jun 21;345(6277):722-4

  (88)   Nishimoto H, Kikutani H, Yamamura K, Kishimoto T. Prevention of autoimmune insulitis by expression of I-E molecules in NOD mice. Nature 1987;328(6129):432-4

  (89)   Wherrett DK, Singer SM, McDevitt HO. Reduction in diabetes incidence in an I-Ag7 transgenic nonobese diabetic mouse line. diab 1997;46(12):1970-4.http://www.ncbi.nlm.nih.gov/pubmed/9392482?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (90)   Ikegami H, Makino S, Harada M, Eisenbarth GS, Hattori M. The cataract Shionogi mouse,a sister non-obese diabetic mouse similar class II but different class I gene products. diabetol 1988;31(4):254-8.http://www.ncbi.nlm.nih.gov/pubmed/3133269?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (91)   Ikegami H, Eisenbarth GS, Hattori M. Major histocompatibility complex-linked diabetogenic gene of the nonobese diabetic mouse. Analysis of genomic DNA amplified by polymerase chain reaction. J Clin Invest 1990;85(1):18-24.http://www.ncbi.nlm.nih.gov/pubmed/2295694?ordinalpos=17&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (92)   Wicker LS, Miller BJ, Fischer PA, Pressey A, Peterson LB. Genetic control of diabetes and insulitis in the nonobese diabetic mouse. Pedigree analysis of a diabetic H-2nod/b heterozygote. J Immunol 1989 Feb 1;142(3):781-4.http://www.ncbi.nlm.nih.gov/pubmed/2643662?ordinalpos=43&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (93)   Corper AL, Stratmann T, Apostolopoulos V, Scott CA, Garcia KC, Kang AS, et al. A structural framework for deciphering the link between I-Ag7 and autoimmune diabetes. Science 2000 Apr 21;288(5465):505-11.http://www.ncbi.nlm.nih.gov/pubmed/10775108?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (94)   Latek RR, Suri A, Petzold SJ, Nelson CA, Kanagawa O, Unanue ER, et al. Structural basis of peptide binding and presentation by the type I diabetes-associated MHC class II molecule of NOD mice. Immunity 2000 Jun;12(6):699-710.http://www.ncbi.nlm.nih.gov/pubmed/10894169?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (95)   Carrasco-Marin E, Kanagawa O, Unanue ER. Insights into the chemistry and biology of the I-Ag7 class II molecule. Res Immunol 1997;148:291-301.http://www.ncbi.nlm.nih.gov/pubmed/9352592?ordinalpos=31&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (96)   Kanagawa O, Martin SM, Vaupel BA, Carrasco-Marin E, Unanue ER. Autoreactivity of T cells from nonobese diabetic mice: an I-Ag7-dependent reaction. Proc Natl Acad Sci U S A 1998 Feb 17;95(4):1721-4.http://www.ncbi.nlm.nih.gov/pubmed/9465083?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (97)   Duarte N, Lundholm M, Holmberg D. The Idd6.2 diabetes susceptibility region controls defective expression of the Lrmp gene in nonobese diabetic (NOD) mice. Immunogenetics 2007 May;59(5):407-16.http://www.ncbi.nlm.nih.gov/pubmed/17353998?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (98)   Schmidt D, Verdaguer J, Averill N, Santamaria P. A mechanism for the major histocompatibility complex-linked resistance to autoimmunity. J Exp Med 1997;186:1059-75.http://www.ncbi.nlm.nih.gov/pubmed/9314555?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

  (99)   Ridgway WM, Ito H, Fasso M, Yu C, Fathman CG. Analysis of the role of variation of major histocompatibility complex class II expression on nonobese diabetic (NOD) peripheral T cell response. J Exp Med 1998 Dec 21;188(12):2267-75

(100)   van Rijn JC, Grote FK, Oostdijk W, Wit JM. Short stature and the probability of coeliac disease, in the absence of gastrointestinal symptoms. Arch Dis Child 2004 Sep;89(9):882-3.http://www.ncbi.nlm.nih.gov/pubmed/15321874?ordinalpos=8&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(101)   Kanagawa O, Vaupel BA, Xu G, Unanue ER, Katz JD. Thymic positive selection and peripheral activation of islet antigen-specific T cells: separation of two diabetogenic steps by an I-A(g7) class II MHC beta-chain mutant. J Immunol 1998 Nov 1;161(9):4489-92.http://www.ncbi.nlm.nih.gov/pubmed/9794372?ordinalpos=18&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(102)   Suri A, Katz JD. Dissecting the role of CD4+ T cells in autoimmune diabetes through the use of TCR transgenic mice. Immunol Rev 1999 Jun;169:55-65.http://www.ncbi.nlm.nih.gov/pubmed/10450508?ordinalpos=8&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(103)   Nakayama M, Beilke JN, Jasinski JM, Kobayashi M, Miao D, Li M, et al. Priming and effector dependence on insulin B:9-23 peptide in NOD islet autoimmunity. J Clin Invest 2007 Jul 2;117(7):1835-43.http://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=ShowDetailView&TermToSearch=17607359&ordinalpos=20&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(104)   Thebault-Baumont K, Dubois-LaForgue D, Krief P, Briand JP, Halbout P, Vallon-Geoffroy K, et al. Acceleration of type 1 diabetes mellitus in proinsulin 2-deficient NOD mice. J Clin Invest 2003 Mar 15;111(6):851-7.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=12639991

(105)   Levisetti MG, Suri A, Petzold SJ, Unanue ER. The insulin-specific T cells of nonobese diabetic mice recognize a weak MHC-binding segment in more than one form. J Immunol 2007 May 15;178(10):6051-7.http://www.ncbi.nlm.nih.gov/pubmed/17475829?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(106)   Michels AW, Nakayama M. 2The anti-insulin trimolecular complex in type 1 diabetes. Curr Opin Endocrinol Diabetes Obes 2010 May 19.PM:20489610

(107)   Kobayashi M, Jasinski J, Liu E, Li M, Miao D, Zhang L, et al. Conserved T cell receptor alpha-chain induces insulin autoantibodies. Proc Natl Acad Sci U S A 2008 Jul 22;105(29):10090-4

(108)   Nikoopour E, Sandrock C, Huszarik K, Krougly O, Lee-Chan E, Masteller EL, et al. Cutting Edge: Vasostatin-1-Derived Peptide ChgA29-42 Is an Antigenic Epitope of Diabetogenic BDC2.5 T Cells in Nonobese Diabetic Mice. J Immunol 2011 Apr 1;186(7):3831-5.PM:21357258

(109)   Ikegami H, Jackson RA, Makino S, Watt DE, Eisenbarth GS, Hattori M. Homozygosity for two genes (H-2: chromosome 17 and Thy-1: chromosome 9) linked to development of type I diabetes of the NOD mouse. Clin Res 34, 683A. 1986.

Ref Type: Abstract

(110)   Lyons PA, Hancock WW, Denny P, Lord CJ, Hill NJ, Armitage N, et al. The NOD Idd9 genetic interval influences the pathogenicity of insulitis and contains molecular variants of Cd30, Tnfr2, and Cd137. Immunity 2000 Jul;13(1):107-15.http://www.ncbi.nlm.nih.gov/pubmed/10933399?ordinalpos=4&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(111)   Podolin PL, Wilusz MB, Cubbon RM, Pajvani U, Lord CJ, Todd JA, et al. Differential glycosylation of interleukin 2, the molecular basis for the NOD Idd3 type 1 diabetes gene? Cytokine 2000 May;12(5):477-82.http://www.ncbi.nlm.nih.gov/pubmed/10857762?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(112)   Eisenbarth SC, Homann D. "Primer Immunology and Autoimmunity". Type I Diabetes: Molecular, Cellular and Clinical Immunology 2006 Nov.http://barbaradaviscenter.org/

(113)   Ikegami H, Fujisawa T, Sakamoto T, Makino S, Ogihara T. Idd1 and Idd3 are necessary but not sufficient for development of type 1 diabetes in NOD mouse. Diabetes Res Clin Pract 2004 Dec;66 Suppl 1:S85-S90.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15563987

(114)   del Rio R, Noubade R, Subramanian M, Saligrama N, Diehl S, Rincon M, et al. SNPs upstream of the minimal promoter control IL-2 expression and are candidates for the autoimmune disease-susceptibility locus Aod2/Idd3/Eae3. Genes Immun 2008 Mar;9(2):115-21.http://www.ncbi.nlm.nih.gov/pubmed/18200031?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(115)   Ghosh S, Palmer SM, Rodrigues NR, Cordell HJ, Hearne CM, Cornall RJ, et al. Polygenic control of autoimmune diabetes in nonobese diabetic mice. Nat Genet 1993;4:404-9.http://www.ncbi.nlm.nih.gov/sites/entrez?orig_db=PubMed&db=pubmed&cmd=Search&term=4%5Bvolume%5D%20AND%20404%5Bpage%5D%20AND%201993%5Bpdat%5D

(116)   McDuffie M, Maybee NA, Keller SR, Stevens BK, Garmey JC, Morris MA, et al. Nonobese diabetic (NOD) mice congenic for a targeted deletion of 12/15-lipoxygenase are protected from autoimmune diabetes. Diabetes 2008 Jan;57(1):199-208.http://www.ncbi.nlm.nih.gov/pubmed/17940120?ordinalpos=121&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(117)   Davoodi-Semiromi A, McDuffie M, Litherland S, Clare-Salzler M. Truncated pStat5B is associated with the Idd4 locus in NOD mice. Biochem Biophys Res Commun 2007 May 11;356(3):655-61.http://www.ncbi.nlm.nih.gov/pubmed/17382905?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(118)   Hill NJ, Lyons PA, Armitage N, Todd JA, Wicker LS, Peterson LB. NOD Idd5 locus controls insulitis and diabetes and overlaps the orthologous CTLA4/IDDM12 and NRAMP1 loci in humans [In Process Citation]. diab 2000 Oct;49(10):1744-7.http://www.ncbi.nlm.nih.gov/pubmed/11016460?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(119)   Fox CJ, Paterson AD, Mortin-Toth SM, Danska JS. Two genetic loci regulate T cell-dependent islet inflammation and drive autoimmune diabetes pathogenesis. Am J Hum Genet 2000 Jul;67(1):67-81.http://www.ncbi.nlm.nih.gov/pubmed/10848492?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(120)   Cordell HJ, Todd JA, Hill NJ, Lord CJ, Lyons PA, Peterson LB, et al. Statistical modeling of interlocus interactions in a complex disease: rejection of the multiplicative model of epistasis in type 1 diabetes. Genetics 2001 May;158(1):357-67.http://www.ncbi.nlm.nih.gov/pubmed/11333244?ordinalpos=19&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(121)   Wicker LS, Chamberlain G, Hunter K, Rainbow D, Howlett S, Tiffen P, et al. Fine mapping, gene content, comparative sequencing, and expression analyses support Ctla4 and Nramp1 as candidates for Idd5.1 and Idd5.2 in the nonobese diabetic mouse. J Immunol 2004 Jul 1;173(1):164-73.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15210771

(122)   Greve B, Vijayakrishnan L, Kubal A, Sobel RA, Peterson LB, Wicker LS, et al. The diabetes susceptibility locus Idd5.1 on mouse chromosome 1 regulates ICOS expression and modulates murine experimental autoimmune encephalomyelitis. J Immunol 2004 Jul 1;173(1):157-63.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15210770

(123)   Araki M, Chung D, Liu S, Rainbow DB, Chamberlain G, Garner V, et al. Genetic evidence that the differential expression of the ligand-independent isoform of CTLA-4 is the molecular basis of the Idd5.1 type 1 diabetes region in nonobese diabetic mice. J Immunol 2009 Oct 15;183(8):5146-57.PM:19783679

(124)   Colucci F, Bergman ML, Penha-Goncalves C, Cilio CM, Holmberg D. Apoptosis resistance of nonobese diabetic peripheral lymphocytes linked to the Idd5 diabetes susceptibility region. Proc Natl Acad Sci U S A 1997 Aug 5;94(16):8670-4.http://www.ncbi.nlm.nih.gov/pubmed/9238035?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(125)   Bergman ML, Cilio CM, Penha-Goncalves C, Lamhamedi-Cherradi SE, Lofgren A, Colucci F, et al. CTLA-4-/- mice display T cell-apoptosis resistance resembling that ascribed to autoimmune-prone non-obese diabetic (NOD) mice. J Autoimmun 2001 Mar;16(2):105-13.http://www.ncbi.nlm.nih.gov/pubmed/11247636?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(126)   Motta V, Lejon K, Holmberg D. The NOD allele of the Idd5 locus on chromosome 1 mediates a non-cell-autonomous defect in negative selection of T cells. J Autoimmun 2007 Jun;28(4):216-23.http://www.ncbi.nlm.nih.gov/pubmed/17449224?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(127)   Irie J, Reck B, Wu Y, Wicker LS, Howlett S, Rainbow D, et al. Genome-wide microarray expression analysis of CD4+ T Cells from nonobese diabetic congenic mice identifies Cd55 (Daf1) and Acadl as candidate genes for type 1 diabetes. J Immunol 2008 Jan 15;180(2):1071-9.http://www.ncbi.nlm.nih.gov/pubmed/18178847?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(128)   Carnaud C, Gombert J, Donnars O, Garchon H, Herbelin A. Protection against diabetes and improved NK/NKT cell performance in NOD.NK1.1 mice congenic at the NK complex. J Immunol 2001 Feb 15;166(4):2404-11.http://www.ncbi.nlm.nih.gov/pubmed/11160299?ordinalpos=5&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(129)   Laloux V, Beaudoin L, Jeske D, Carnaud C, Lehuen A. NK T cell-induced protection against diabetes in V alpha 14-J alpha 281 transgenic nonobese diabetic mice is associated with a Th2 shift circumscribed regionally to the islets and functionally to islet autoantigen. J Immunol 2001 Mar 15;166(6):3749-56.http://www.ncbi.nlm.nih.gov/pubmed/11238616?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(130)   Sharif S, Arreaza GA, Zucker P, Mi QS, Sondhi J, Naidenko OV, et al. Activation of natural killer T cells by alpha-galactosylceramide treatment prevents the onset and recurrence of autoimmune Type 1 diabetes. Nat Med 2001 Sep;7(9):1057-62.http://www.ncbi.nlm.nih.gov/pubmed/11533711?ordinalpos=4&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(131)   Bergman ML, Penha-Goncalves C, Lejon K, Holmberg D. Low rate of proliferation in immature thymocytes of the non-obese diabetic mouse maps to the Idd6 diabetes susceptibility region. diabetol 2001 Aug;44(8):1054-61.http://www.ncbi.nlm.nih.gov/pubmed/11484085?ordinalpos=24&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(132)   Vallois D, Grimm CH, Avner P, Boitard C, Rogner UC. The type 1 diabetes locus Idd6 controls TLR1 expression. J Immunol 2007 Sep 15;179(6):3896-903.http://www.ncbi.nlm.nih.gov/pubmed/17785827?ordinalpos=156&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(133)   Serreze DV, Choisy-Rossi CM, Grier AE, Holl TM, Chapman HD, Gahagan JR, et al. Through regulation of TCR expression levels, an Idd7 region gene(s) interactively contributes to the impaired thymic deletion of autoreactive diabetogenic CD8+ T cells in nonobese diabetic mice. J Immunol 2008 Mar 1;180(5):3250-9.http://www.ncbi.nlm.nih.gov/pubmed/18292549?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(134)   Siegmund T, Armitage N, Wicker LS, Peterson LB, Todd JA, Lyons PA. Analysis of the mouse CD30 gene: a candidate for the NOD mouse type 1 diabetes locus Idd9.2. diab 2000 Sep;49(9):1612-6.http://www.ncbi.nlm.nih.gov/pubmed/10969850?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(135)   Ueno A, Wang J, Cheng L, Im JS, Shi Y, Porcelli SA, et al. Enhanced early expansion and maturation of semi-invariant NK T cells inhibited autoimmune pathogenesis in congenic nonobese diabetic mice. J Immunol 2008 Nov 15;181(10):6789-96.PM:18981096

(136)   Rolf J, Motta V, Duarte N, Lundholm M, Berntman E, Bergman ML, et al. The enlarged population of marginal zone/CD1d(high) B lymphocytes in nonobese diabetic mice maps to diabetes susceptibility region Idd11. Journal of Immunology 2005;174(8):4821-7.<Go to ISI>://000228234600048

(137)   Rolf J, Motta V, Duarte N, Lundholm M, Berntman E, Bergman ML, et al. Congenic nonobese diabetic mouse strains fail to confirm linkage of a marginal zone B lymphocyte phenotype to the Idd11 locus on chromosome 4 - Response. Journal of Immunology 2006;176(2):702.<Go to ISI>://000234553800002

(138)   Heath VL, Moore NC, Parnell SM, Mason DW. Intrathymic expression of genes involved in organ specific autoimmune disease. J Autoimmun 1998 Aug;11(4):309-18.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=9776708

(139)   Yamaji K, Ikegami H, Fujisawa T, Noso S, Nojima K, Babaya N, et al. Evidence for Cd101 but not Fcgr1 as candidate for type 1 diabetes locus, Idd10. Biochem Biophys Res Commun 2005 Jun 3;331(2):536-42.http://www.ncbi.nlm.nih.gov/pubmed/15850792?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(140)   Podolin PL, Denny P, Lord CJ, Hill NJ, Todd JA, Peterson LB, et al. Congenic mapping of the insulin-dependent diabetes (Idd) gene, Idd10, localizes two genes mediating the Idd10 effect and eliminates the candidate Fcgr1. J Immunol 1997 Aug 15;159(4):1835-43.http://www.ncbi.nlm.nih.gov/pubmed/9257847?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(141)   Chen YG, Driver JP, Silveira PA, Serreze DV. Subcongenic analysis of genetic basis for impaired development of invariant NKT cells in NOD mice. Immunogenetics 2007;59(9):705-12.<Go to ISI>://000249503200002

(142)   Schmidt MB. Eine biglandulare Erkrankung (Nebennieren und Schilddruse) bei Morbus Addisonii. Verh Dtsch Ges Pathol 1926;21:212-21

(143)   Liston A, Gray DH, Lesage S, Fletcher AL, Wilson J, Webster KE, et al. Gene dosage--limiting role of Aire in thymic expression, clonal deletion, and organ-specific autoimmunity. J Exp Med 2004 Oct 18;200(8):1015-26.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15492124

(144)   Anderson MS. Autoimmune endocrine disease. Curr Opin Immunol 2002 Dec;14(6):760-4.http://www.ncbi.nlm.nih.gov/pubmed/12413526?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(145)   Adams TE, Apert S, Hanahan D. Non-tolerance and autoantibodies to a transgenic self-antigen expressed in pancreatic beta cells. Nature 1987;325:223-8.http://www.ncbi.nlm.nih.gov/pubmed/3543686?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(146)   Hanahan D. Peripheral-antigen-expressing cells in thymic medulla: factors in self- tolerance and autoimmunity. Curr Opin Immunol 1998 Dec;10(6):656-62.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=9914224

(147)   Pugliese A. Genetics of type 1 diabetes. Endocrinol Metab Clin North Am 2004 Mar;33(1):1-16, vii.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15053891

(148)   Cetani F, Barbesino G, Borsari S, Pardi E, Cianferotti L, Pinchera A, et al. A novel mutation of the autoimmune regulator gene in an Italian kindred with autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy, acting in a dominant fashion and strongly cosegregating with hypothyroid autoimmune thyroiditis. J Clin Endocrinol Metab 2001 Oct;86(10):4747-52.http://www.ncbi.nlm.nih.gov/pubmed/11600535?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(149)   Gray DH, Gavanescu I, Benoist C, Mathis D. Danger-free autoimmune disease in Aire-deficient mice. Proc Natl Acad Sci U S A 2007 Nov 13;104(46):18193-8.http://www.ncbi.nlm.nih.gov/pubmed/17991771?ordinalpos=102&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(150)   Gambineri E, Torgerson TR, Ochs HD. Immune dysregulation, polyendocrinopathy, enteropathy, and X-linked inheritance (IPEX), a syndrome of systemic autoimmunity caused by mutations of FOXP3, a critical regulator of T-cell homeostasis. Current Opinion in Rheumatology 2003;15(4):430-5.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=12819471

(151)   Patel DD. Escape from tolerance in the human X-linked autoimmunity-allergic disregulation syndrome and the Scurfy mouse. J Clin Invest 2001;107(2):155-7.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=11160129&query_hl=7

(152)   Chen Z, Benoist C, Mathis D. How defects in central tolerance impinge on a deficiency in regulatory T cells. Proc Natl Acad Sci U S A 2005 Oct 11;102(41):14735-40.http://www.ncbi.nlm.nih.gov/pubmed/16203996?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(153)   Griffiths MM, Encinas JA, Remmers EF, Kuchroo VK, Wilder RL. Mapping autoimmunity genes. Curr Opin Immunol 1999 Dec;11(6):689-700.http://www.ncbi.nlm.nih.gov/pubmed/10631556?ordinalpos=4&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(154)   Fairhurst AM, Wandstrat AE, Wakeland EK. Systemic lupus erythematosus: multiple immunological phenotypes in a complex genetic disease. Adv Immunol 2006;92:1-69.:1-69.http://www.ncbi.nlm.nih.gov/pubmed/17145301?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(155)   Bottini N, Musumeci L, Alonso A, Rahmouni S, Nika K, Rostamkhani M, et al. A functional variant of lymphoid tyrosine phosphatase is associated with type I diabetes. Nat Genet 2004 Apr;36(4):337-8.http://www.ncbi.nlm.nih.gov/sites/entrez?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=15004560

(156)   Smyth D, Cooper JD, Collins JE, Heward JM, Franklyn JA, Howson JM, et al. Replication of an Association Between the Lymphoid Tyrosine Phosphatase Locus (LYP/PTPN22) With Type 1 Diabetes, and Evidence for Its Role as a General Autoimmunity Locus. diab 2004 Nov;53(11):3020-3.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15504986

(157)   Velaga MR, Wilson V, Jennings CE, Owen CJ, Herington S, Donaldson PT, et al. The Codon 620 Tryptophan Allele of the Lymphoid Tyrosine Phosphatase (LYP) Gene Is a Major Determinant of Graves' Disease. J Clin Endocrinol Metab 2004 Dec;89(11):5862-5.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15531553

(158)   Salomon B, Rhee L, Bour-Jordan H, Hsin H, Montag A, Soliven B, et al. Development of spontaneous autoimmune peripheral polyneuropathy in b7-2-deficient nod mice. J Exp Med 2001 Sep 3;194(5):677-84.http://www.ncbi.nlm.nih.gov/pubmed/11535635?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(159)   Winer S, Tsui H, Lau A, Song A, Li X, Cheung RK, et al. Autoimmune islet destruction in spontaneous type 1 diabetes is not beta-cell exclusive. Nat Med 2003 Feb;9(2):198-205.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=12539039

(160)   Winer S, Astsaturov I, Cheung R, Gunaratnam L, Kubiak V, Cortez MA, et al. Type I diabetes and multiple sclerosis patients target islet plus central nervous system autoantigens; nonimmunized nonobese diabetic mice can develop autoimmune encephalitis. J Immunol 2001 Feb 15;166(4):2831-41.http://www.ncbi.nlm.nih.gov/pubmed/11160351?ordinalpos=8&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(161)   Rivero VE, Cailleau C, Depiante-Depaoli M, Riera CM, Carnaud C. Non-obese diabetic (NOD) mice are genetically susceptible to experimental autoimmune prostatitis (EAP). J Autoimmun 1998 Dec;11(6):603-10.http://www.ncbi.nlm.nih.gov/pubmed/9878082?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(162)   Koarada S, Wu Y, Fertig N, Sass DA, Nalesnik M, Todd JA, et al. Genetic control of autoimmunity: protection from diabetes, but spontaneous autoimmune biliary disease in a nonobese diabetic congenic strain. J Immunol 2004 Aug 15;173(4):2315-23.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15294944

(163)   Bendelac A, Carnaud C, Boitard C, Bach JF. Syngeneic transfer of autoimmune diabetes from diabetic NOD mice to healthy neonates.  Requirement for both L3T4+ and Lyt-2+ T cells. J Exp Med 1987;166:823-32.http://www.ncbi.nlm.nih.gov/pubmed/3309126?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(164)   Wicker LS, Miller BJ, Mullen Y. Transfer of autoimmune diabetes mellitus with splenocytes from non-obese diabetic (NOD) mice. diab 1986;35(8):855-60.http://www.ncbi.nlm.nih.gov/pubmed/3525284?ordinalpos=7&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(165)   Charlton B, Mandel TE. Progression from insulitis to Beta cell destruction in NOD mouse requires L3T4+T-lymphocytes. diab 1988;37:1108-12.http://www.ncbi.nlm.nih.gov/pubmed/3292330?ordinalpos=7&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(166)   Serreze DV, Chapman HD, Varnum DS, Hanson MS, Reifsnyder PC, Scott DR, et al. B lymphocytes are essential for the initiation of T cell-mediated autoimmune diabetes: analysis of a new "speed-congenic" stock of NOD.Igunull mice. J Exp Med 1996;184:2049-53.http://www.ncbi.nlm.nih.gov/pubmed/8920894?ordinalpos=11&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(167)   Greeley SA, Katsumata M, Yu L, Eisenbarth GS, Moore DJ, Goodarzi H, et al. Elimination of maternally transmitted autoantibodies prevents diabetes in nonobese diabetic mice. Nat Med 2002 Apr;8(4):399-402.http://www.ncbi.nlm.nih.gov/pubmed/11927947?ordinalpos=4&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(168)   Honkanen J, Nieminen JK, Gao R, Luopajarvi K, Salo HM, Ilonen J, et al. IL-17 immunity in human type 1 diabetes. J Immunol 2010 Aug 1;185(3):1959-67.PM:20592279

(169)   Sreenan S, Pick AJ, Levisetti M, Baldwin AC, Pugh W, Polonsky KS. Increased b-cell proliferation and reduced mass before diabetes onset in the nonobese diabetic mouse. diab 1999;48:989-96.http://www.ncbi.nlm.nih.gov/pubmed/10331402?ordinalpos=20&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(170)   Valorani MG, Hawa MI, Buckley LR, Afeltra A, Cacciapaglia F, Pozzilli P. The natural history of insulin content in the pancreas of female and male non-obese diabetic mouse: implications for trials of diabetes prevention in humans. Diabetes Metab Res Rev 2004 Sep;20(5):394-8.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15343585

(171)   Alanentalo T, Hornblad A, Mayans S, Karin NA, Sharpe J, Larefalk A, et al. Quantification and three-dimensional imaging of the insulitis-induced destruction of beta-cells in murine type 1 diabetes. diab 2010 Jul;59(7):1756-64.PM:20393145

(172)   Nir T, Melton DA, Dor Y. Recovery from diabetes in mice by beta cell regeneration. J Clin Invest 2007 Sep;117(9):2553-61.http://www.ncbi.nlm.nih.gov/sites/entrez?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=17786244

(173)   Meier JJ, Lin JC, Butler AE, Galasso R, Martinez DS, Butler PC. Direct evidence of attempted beta cell regeneration in an 89-year-old patient with recent-onset type 1 diabetes. Diabetologia 2006 Aug;49(8):1838-44.http://www.ncbi.nlm.nih.gov/pubmed/16802132?ordinalpos=9&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(174)   Butler AE, Galasso R, Meier JJ, Basu R, Rizza RA, Butler PC. Modestly increased beta cell apoptosis but no increased beta cell replication in recent-onset type 1 diabetic patients who died of diabetic ketoacidosis. diabetol 2007 Nov;50(11):2323-31.http://www.ncbi.nlm.nih.gov/pubmed/17805509?dopt=Citation

(175)   Perl S, Kushner JA, Buchholz BA, Meeker AK, Stein GM, Hsieh M, et al. Significant human beta-cell turnover is limited to the first three decades of life as determined by in vivo thymidine analog incorporation and radiocarbon dating. J Clin Endocrinol Metab 2010 Oct;95(10):E234-E239.PM:20660050

(176)   Keenan HA, Sun JK, Levine J, Doria A, Aiello LP, Eisenbarth G, et al. Residual insulin production and pancreatic ss-cell turnover after 50 years of diabetes: Joslin Medalist Study. diab 2010 Nov;59(11):2846-53.PM:20699420

(177)   Sherry NA, Kushner JA, Glandt M, Kitamura T, Brillantes AM, Herold KC. Effects of autoimmunity and immune therapy on beta-cell turnover in type 1 diabetes. Diabetes 2006 Dec;55(12):3238-45.http://www.ncbi.nlm.nih.gov/pubmed/17130466?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(178)   Haskins K, Atkinson M. Prediction of Diabetes and Response to Therapy in Individual Animals: are there Lessons for Man? Report on the JDF/Snow Mountain Meeting, 5-7 October 2000, Winter Park, CO, USA. J Autoimmun 2001 Feb;16(1):15-9.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=11221992

(179)   Bonifacio E, Atkinson M, Eisenbarth G, Serreze D, Kay TW, Lee-Chan E, et al. International Workshop on Lessons from Animal Models for Human Type 1 Diabetes: analyzing target autoantigens of humoral immunity in nonobese diabetic mice. Ann N Y Acad Sci 2002 Apr;958:1-2.http://www.ncbi.nlm.nih.gov/pubmed/12021078?ordinalpos=4&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(180)   Yu L, Eisenbarth GS, Bonifacio E, Thomas J, Atkinson M, Wasserfall C. The Second Murine Autoantibody Workshop: remarkable inter-laboratory concordance for radiobinding assays to identify insulin autoantibodies in non-obese diabetic mice. NY Acad Sci 2003;100:1-12.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=14679035

(181)   Moore DJ, Noorchashm H, Lin TH, Greeley SA, Naji A. NOD B-cells are insufficient to incite T-cell-mediated anti-islet autoimmunity. Diabetes 2005 Jul;54(7):2019-25.http://www.ncbi.nlm.nih.gov/pubmed/15983202?ordinalpos=12&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(182)   Noorchashm H, Noorchashm N, Kern J, Rostami SY, Barker CF, Naji A. B-cells are required for the initiation of insulitis and sialitis in nonobese diabetic mice. diab 1997 Jun;46(6):941-6.http://www.ncbi.nlm.nih.gov/pubmed/9166663?ordinalpos=5&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(183)   Kendall PL, Yu G, Woodward EJ, Thomas JW. Tertiary lymphoid structures in the pancreas promote selection of B lymphocytes in autoimmune diabetes. J Immunol 2007 May 1;178(9):5643-51.http://www.ncbi.nlm.nih.gov/pubmed/17442947?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(184)   Hu CY, Rodriguez-Pinto D, Du W, Ahuja A, Henegariu O, Wong FS, et al. Treatment with CD20-specific antibody prevents and reverses autoimmune diabetes in mice. J Clin Invest 2007 Dec;117(12):3857-67.http://www.ncbi.nlm.nih.gov/pubmed/18060033?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(185)   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 Jan 18;189(2):331-9.http://www.ncbi.nlm.nih.gov/pubmed/9892615?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(186)   Denis MC, Mahmood U, Benoist C, Mathis D, Weissleder R. Imaging inflammation of the pancreatic islets in type 1 diabetes. Proc Natl Acad Sci U S A 2004 Aug 24;101(34):12634-9.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15304647

(187)   Lejon K, Fathman CG. Isolation of self antigen-reactive cells from inflamed islets of nonobese diabetic mice using CD4high expression as a marker. J Immunol 1999 Nov 15;163(10):5708-14.http://www.ncbi.nlm.nih.gov/pubmed/10553102?ordinalpos=17&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(188)   Gagnerault MC, Luan JJ, Lotton C, Lepault F. Pancreatic lymph nodes are required for priming of beta cell reactive T cells in NOD mice. J Exp Med 2002 Aug 5;196(3):369-77.http://www.ncbi.nlm.nih.gov/pubmed/12163565?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(189)   Poirot L, Benoist C, Mathis D. Natural killer cells distinguish innocuous and destructive forms of pancreatic islet autoimmunity. Proc Natl Acad Sci U S A 2004 May 25;101(21):8102-7.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15141080

(190)   Serreze DV, Gaskins HR, Leiter EH. Defects in the differentiation and function of antigen presenting cells in NOD/Lt mice. J Immunol 1993 Mar 15;150(6):2534-43.http://www.ncbi.nlm.nih.gov/pubmed/8450229?ordinalpos=4&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(191)   Piganelli JD, Martin T, Haskins K. Splenic macrophages from the NOD mouse are defective in the ability to present antigen. diab 1998;47:1212-8.http://www.ncbi.nlm.nih.gov/pubmed/9703319?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(192)   Dahlen E, Hedlund G, Dawe K. Low CD86 expression in the nonobese diabetic mouse results in the impairment of both T cell activation and CTLA-4 up-regulation. J Immunol 2000 Mar 1;164(5):2444-56.http://www.ncbi.nlm.nih.gov/pubmed/10679081?ordinalpos=29&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(193)   Strid J, Lopes L, Marcinkiewicz J, Petrovska L, Nowak B, Chain BM, et al. A defect in bone marrow derived dendritic cell maturation in the nonobesediabetic mouse. Clin Exp Immunol 2001 Mar;123(3):375-81.http://www.ncbi.nlm.nih.gov/pubmed/11298122?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(194)   Wong FS, Karttunen J, Dumont C, Wen L, Visintin I, Pilip IM, et al. Identification of an MHC class I-restricted autoantigen in type 1 diabetes by screening an organ-specific cDNA library. Nat Med 1999 Sep;5(9):1026-31.http://www.ncbi.nlm.nih.gov/pubmed/10470079?ordinalpos=4&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(195)   Buchs AE, Rapoport MJ. T cell signaling and autoimmune diabetes. J Pediatr Endocrinol Metab 2000 Nov;13(9):1549-54.http://www.ncbi.nlm.nih.gov/pubmed/11154149?ordinalpos=6&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(196)   Zhang J, Salojin KV, Delovitch TL. CD28 co-stimulation restores T cell responsiveness in NOD mice by overcoming deficiencies in Rac-1/p38 mitogen-activated protein kinase signaling and IL-2 and IL-4 gene transcription. Int Immunol 2001 Mar;13(3):377-84.http://www.ncbi.nlm.nih.gov/pubmed/11222507?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(197)   King C, Ilic A, Koelsch K, Sarvetnick N. Homeostatic expansion of T cells during immune insufficiency generates autoimmunity. Cell 2004 Apr 16;117(2):265-77.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15084263

(198)   Lang J, Bellgrau D. A T-cell functional phenotype common among autoimmune-prone rodent strains. Scand J Immunol 2002 Jun;55(6):546-59.http://www.ncbi.nlm.nih.gov/pubmed/12028557?ordinalpos=21&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(199)   McInerney MF, Pek SB, Thomas DW. Prevention of insulitis and diabetes onset by treatment with complete Freund's adjuvant in NOD mice. diab 1991;40(6):715-25.http://www.ncbi.nlm.nih.gov/pubmed/2040388?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(200)   Guytingco R, Quddus J, Richardson B, Pek SB. Single administration of bacillus Calmette-Guerin (BCG) vaccine in nonobese diabetic (NOD) mice results in suppression of in vitro T-lymphocyte proliferation and regression of insulitis. Diabetes 40, 152A. 1991.

Ref Type: Abstract

(201)   Singh B. Stimulation of the developing immune system can prevent autoimmunity. J Autoimmun 2000 Feb;14(1):15-22.http://www.ncbi.nlm.nih.gov/pubmed/10648113?ordinalpos=13&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(202)   Ridgway WM, Fassò M, Lanctot A, Garvey C, Fathman CG. Breaking self-tolerance in nonobese diabetic mice. J Exp Med 1996;183:1657-62.http://www.ncbi.nlm.nih.gov/pubmed/8666923?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(203)   Boyton RJ, Lohmann T, Londei M, Kalbacher H, Halder T, Frater AJ, et al. Glutamic acid decarboxylase T lymphocyte responses associated with susceptibility or resistance to type I diabetes: analysis in disease discordant human twins, non-obese diabetic mice and HLA-DQ transgenic mice. Int Immunol 1998 Dec;10(12):1765-76.http://www.ncbi.nlm.nih.gov/pubmed/9885897?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(204)   Dosch HM, Becker DJ. Measurement of T-cell autoreactivity in autoimmune diabetes [letter] [In Process Citation]. diabetol 2000 Mar;43(3):386-7.http://www.ncbi.nlm.nih.gov/pubmed/10768103?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(205)   Kreuwel HT, Biggs JA, Pilip IM, Pamer EG, Lo D, Sherman LA. Defective CD8+ T cell peripheral tolerance in nonobese diabetic mice. J Immunol 2001 Jul 15;167(2):1112-7.http://www.ncbi.nlm.nih.gov/pubmed/11441123?ordinalpos=11&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(206)   Tian J, Gregori S, Adorini L, Kaufman DL. The frequency of high avidity T cells determines the hierarchy of determinant spreading. J Immunol 2001 Jun 15;166(12):7144-50.http://www.ncbi.nlm.nih.gov/pubmed/11390460?ordinalpos=139&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(207)   Hutton JC, Eisenbarth GS. A pancreatic {beta}-cell-specific homolog of glucose-6-phosphatase emerges as a major target of cell-mediated autoimmunity in diabetes. Proc Natl Acad Sci U S A 2003 Jul 14;100:8626-8.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=pubmed&cmd=Retrieve&dopt=AbstractPlus&list_uids=12861077

(208)   Trudeau JD, Kelly-Smith C, Verchere CB, Elliott JF, Dutz JP, Finegood DT, et al. Prediction of spontaneous autoimmune diabetes in NOD mice by quantification of autoreactive T cells in peripheral blood. J Clin Invest 2003 Jan;111(2):217-23.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=12531877

(209)   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 Aug 17;406(6797):739-42.http://www.ncbi.nlm.nih.gov/pubmed/10963600?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(210)   Naquet P, Naspetti M, Boyd R. Development, organization and function of the thymic medulla in normal, immunodeficient or autoimmune mice. Semin Immunol 1999 Feb;11(1):47-55.http://www.ncbi.nlm.nih.gov/pubmed/9950751?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(211)   Lord CJ, Howlett S, Lyons PA, Peterson LB, Wicker LS, Todd JA. The murine type 1 diabetes loci, Idd1, Idd3, Idd5, Idd9, and Idd17/10/18, do not control thymic CD4-CD8-/TCRalphabeta+ deficiency in the nonobese diabetic mouse. Mamm Genome 2001 Feb;12(2):175-6.http://www.ncbi.nlm.nih.gov/pubmed/11210190?ordinalpos=25&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(212)   Bluestone JA, Tang Q. Therapeutic vaccination using CD4+CD25+ antigen-specific regulatory T cells. Proc Natl Acad Sci U S A 2004 Oct 5;101 Suppl 2:14622-6.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15322272

(213)   Tang Q, Bluestone JA. The Foxp3+ regulatory T cell: a jack of all trades, master of regulation. Nat Immunol 2008 Mar;9(3):239-44.http://www.ncbi.nlm.nih.gov/pubmed/18285775?ordinalpos=6&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(214)   Masteller EL, Warner MR, Tang Q, Tarbell KV, McDevitt H, Bluestone JA. Expansion of functional endogenous antigen-specific CD4+CD25+ regulatory T cells from nonobese diabetic mice. J Immunol 2005 Sep 1;175(5):3053-9.http://www.ncbi.nlm.nih.gov/pubmed/16116193?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(215)   Hutchings PR, Cooke A. The transfer of autoimmune diabetes in NOD mice can be inhibited or accelerated by distinct cell populations present in normal splenocytes taken from young males. J Autoimmun 1990 Apr;3(2):175-85.http://www.ncbi.nlm.nih.gov/pubmed/1971173?ordinalpos=9&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(216)   Boitard C, Yasunami R, Dardenne M, Bach JF. T cell-mediated inhibition of the transfer of autoimmune diabetes in NOD mice. J Exp Med 1989 May 1;169(5):1669-80.http://www.ncbi.nlm.nih.gov/pubmed/2523954?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(217)   Belghith M, Bluestone JA, Barriot S, Megret J, Bach JF, Chatenoud L. TGF-beta-dependent mechanisms mediate restoration of self-tolerance induced by antibodies to CD3 in overt autoimmune diabetes. Nat Med 2003 Sep;9(9):1202-8.http://www.ncbi.nlm.nih.gov/pubmed/12937416?ordinalpos=4&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(218)   Weiner HL. Oral tolerance, an active immunologic process mediated by multiple mechanisms. J Clin Invest 2000 Oct;106(8):935-7.http://www.ncbi.nlm.nih.gov/pubmed/11032852?ordinalpos=13&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(219)   Roncarolo MG, Gregori S. Is FOXP3 a bona fide marker for human regulatory T cells? Eur J Immunol 2008 Apr;38(4):925-7.http://www.ncbi.nlm.nih.gov/pubmed/18395862?ordinalpos=8&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(220)   Trucco M, Giannoukakis N. Immunoregulatory dendritic cells to prevent and reverse new-onset Type 1 diabetes mellitus. Expert Opin Biol Ther 2007 Jul;7(7):951-63.http://www.ncbi.nlm.nih.gov/pubmed/17665986?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(221)   Du W, Wong FS, Li MO, Peng J, Qi H, Flavell RA, et al. TGF-beta signaling is required for the function of insulin-reactive T regulatory cells. J Clin Invest 2006 May 1;116(5):1360-70.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=pubmed&cmd=Retrieve&dopt=AbstractPlus&list_uids=16670772

(222)   Pop SM, Wong CP, He Q, Wang Y, Wallet MA, Goudy KS, et al. The type and frequency of immunoregulatory CD4+ T-cells govern the efficacy of antigen-specific immunotherapy in nonobese diabetic mice. Diabetes 2007 May;56(5):1395-402.http://www.ncbi.nlm.nih.gov/pubmed/17317763?ordinalpos=8&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(223)   Fehervari Z, Sakaguchi S. CD4(+) Tregs and immune control. J Clin Invest 2004 Dec;114(9):1209-17.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15520849

(224)   Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science 2003 Feb 14;299(5609):1057-61.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=12522256

(225)   Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 2003 Apr;4(4):330-6.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=12612578

(226)   Jaeckel E, von Boehmer H, Manns MP. Antigen-Specific FoxP3-Transduced T-Cells Can Control Established Type 1 Diabetes. diab 2005;54(2):306-10.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15591438

(227)   Mchugh RS, Shevach EM. Cutting edge: Depletion of CD4(+)CD25(+) regulatory T cells is necessary, but not sufficient, for induction of organ-specific autoimmune disease. Journal of Immunology 2002;168(12):5979-83.http://www.ncbi.nlm.nih.gov/pubmed/12055202?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(228)   Peng Y, Laouar Y, Li MO, Green EA, Flavell RA. TGF-beta regulates in vivo expansion of Foxp3-expressing CD4+CD25+ regulatory T cells responsible for protection against diabetes. Proc Natl Acad Sci U S A 2004 Mar 30;101(13):4572-7.http://www.ncbi.nlm.nih.gov/pubmed/15070759?ordinalpos=6&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(229)   Tang Q, Henriksen KJ, Bi M, Finger EB, Szot G, Ye J, et al. In vitro-expanded antigen-specific regulatory T cells suppress autoimmune diabetes. J Exp Med 2004 Jul 7;199(11):1455-65.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15184499

(230)   Lenschow DJ, Herold KC, Rhee L, Patel B, Koons A, Qin HY, et al. CD28/B7 regulation of Th1 and Th2 subsets in the development of autoimmune diabetes [published erratum appears in Immunity 1997 Feb;6(2):following 215]. Immunity 1996 Sep;5(3):285-93.http://www.ncbi.nlm.nih.gov/pubmed/8808683?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(231)   Herman AE, Freeman GJ, Mathis D, Benoist C. CD4+CD25+ T regulatory cells dependent on ICOS promote regulation of effector cells in the prediabetic lesion. J Exp Med 2004 Jun 7;199(11):1479-89.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15184501

(232)   Mukherjee R, Chaturvedi P, Qin HY, Singh B. CD4+CD25+ regulatory T cells generated in response to insulin B:9-23 peptide prevent adoptive transfer of diabetes by diabetogenic T cells. J Autoimmun 2003 Nov;21(3):221-37.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=14599847

(233)   Harrison LC, Solly NR, Martinez NR. (Pro)insulin-specific regulatory T cells. Novartis Found Symp 2003;252:132-41.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=14609216

(234)   Gombert JM, Herbelin A, Tancrede-Bohin E, Dy M, Carnaud C, Bach JF. Early quantitative and functional deficiency of NK1+-like thymocytes in the NOD mouse. Eur J Immunol 1996;26(12):2989-98.http://www.ncbi.nlm.nih.gov/pubmed/8977295?ordinalpos=6&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(235)   Falcone M, Yeung B, Tucker L, Rodriguez E, Sarvetnick N. A defect in interleukin 12-induced activation and interferon gamma secretion of peripheral natural killer T cells in nonobese diabetic mice suggests new pathogenic mechanisms for insulin-dependent diabetes mellitus. J Exp Med 1999 Oct 4;190(7):963-72.http://www.ncbi.nlm.nih.gov/pubmed/10510086?ordinalpos=5&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(236)   Lee IF, Qin H, Trudeau J, Dutz J, Tan R. Regulation of autoimmune diabetes by complete Freund's adjuvant is mediated by NK cells. J Immunol 2004 Jan 15;172(2):937-42.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=14707066

(237)   Homann D, Jahreis A, Wolfe T, Hughes A, Coon B, van Stipdonk MJ, et al. CD40L blockade prevents autoimmune diabetes by induction of bitypic NK/DC regulatory cells. Immunity 2002 Mar;16(3):403-15.http://www.ncbi.nlm.nih.gov/pubmed/11911825?ordinalpos=9&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(238)   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(8):1853-7.http://www.ncbi.nlm.nih.gov/pubmed/8056042?ordinalpos=7&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(239)   Lieberman SM, Evans AM, Han B, Takaki T, Vinnitskaya Y, Caldwell JA, et al. Identification of the {beta} cell antigen targeted by a prevalent population of pathogenic CD8+ T cells in autoimmune diabetes. Proc Natl Acad Sci U S A 2003 Jul 8;100(14):8384-8.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=12815107

(240)   Lieberman SM, DiLorenzo TP. A comprehensive guide to antibody and T-cell responses in type 1 diabetes. Tissue Antigens 2003 Nov;62(5):359-77.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=14617043

(241)   Li SW, Koya V, Li Y, Donelan W, Lin P, Reeves WH, et al. Pancreatic duodenal homeobox 1 protein is a novel beta-cell-specific autoantigen for type I diabetes. Lab Invest 2010 Jan;90(1):31-9.PM:19901909

(242)   Gurr W, Shaw M, Li Y, Sherwin R. RegII is a beta-cell protein and autoantigen in diabetes of NOD mice. Diabetes 2007 Jan;56(1):34-40

(243)   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 Jun 16;197(12):1635-44.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=12796471

(244)   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 Dec 1;173(11):6727-34.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15557165

(245)   Cohen IR. Thoughts on the pathogenesis of type 1 diabetes and on the arrest of autoimmune beta-cell destruction by peptide p277 vaccination. Isr Med Assoc J 2004 May;6(5):260-1.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15151361

(246)   Bonifacio E, Atkinson M, Eisenbarth GS, Serreze D, Kay TW, Lee-Chan E, et al. International workshop on autoimmunity in animal models of autoimmune diabetes identifies insulin, but not GAD or IA-2 as specific antigens of humoral autoimmunity in the non-obese diabetic mouse. diab 2001;50:2451-8.http://www.ncbi.nlm.nih.gov/pubmed/11679421?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(247)   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(4):1056-62.http://www.ncbi.nlm.nih.gov/pubmed/7537670?ordinalpos=15&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(248)   Jaeckel E, Lipes MA, von Boehmer H. Recessive tolerance to preproinsulin 2 reduces but does not abolish type 1 diabetes. Nat Immunol 2004 Oct;5(10):1028-35.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15378058

(249)   Narendran P, Mannering SI, Harrison LC. Proinsulin - a pathogenic autoantigen in type 1 diabetes. Autoimmunity Reviews 2003;2(4):204-10.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=12848947

(250)   Muir A, Luchetta R, Song H-Y, Peck A, Krischer J, Maclaren N. Insulin immunization protects NOD mice from diabetes. Autoimmunity 15, 58. 1993.

Ref Type: Abstract

(251)   Muir A, Peck A, Clare-Salzler M, Song Y-H, Cornelius J, Luchetta R, et al. Insulin immunization of nonobese diabetic mice induces a protective insulitis characterized by diminished intraislet interferon-gamma transcription. J Clin Invest 1995;95(2):628-34.http://www.ncbi.nlm.nih.gov/pubmed/7860747?ordinalpos=25&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(252)   Hancock WW, Polansky M, Zhang J, Blogg N, Weiner HL. Suppression of insulitis in non-obese diabetic (NOD) mice by oral insulin administration is associated with selective expression of interleukin-4 and -10, transforming growth factor-beta, and prostaglandin-E. Am J Pathol 1995;147(5):1193-9.http://www.ncbi.nlm.nih.gov/pubmed/7485382?ordinalpos=30&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(253)   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 U S A 1991 Nov 15;88(22):10252-6.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=1946445

(254)   Simone E, Daniel D, Schloot N, Gottlieb P, Babu S, Kawasaki E, et al. T cell receptor restriction of diabetogenic autoimmune NOD T cells. Proc Natl Acad Sci USA 1997;94(6):2518-21

(255)   Simone EA, Yu L, Wegmann DR, Eisenbarth GS. T cell receptor gene polymorphisms associated with anti-insulin, autoimmune T cells in diabetes-prone NOD mice. J Autoimmun 1997;10:317-21

(256)   Abiru N, Wegmann D, Kawasaki E, Gottlieb P, Simone E, Eisenbarth GS. Dual overlapping peptides recognized by insulin peptide B:9-23 reactive T cell receptor AV13S3 T cell clones of the NOD mouse. J Autoimmun 2000;14(3):231-7

(257)   Daniel D, Wegmann DR. Protection of nonobese diabetic mice from diabetes by intranasal or subcutaneous administration of insulin peptide B-(9-23). Proc Natl Acad Sci USA 1996;93(2):956-60.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=pubmed&cmd=Retrieve&dopt=AbstractPlus&list_uids=8570667&query_hl=58&itool=pubmed_docsum

(258)   Wong FS, Janeway CAJ. The role of CD4 vs. CD8 T cells in IDDM. J Autoimmun 1999 Nov;13(3):290-5

(259)   Tsai S, Shameli A, Yamanouchi J, Clemente-Casares X, Wang J, Serra P, et al. Reversal of autoimmunity by boosting memory-like autoregulatory T cells. Immunity 2010 Apr 23;32(4):568-80.PM:20381385

(260)   Kash SF, Condie BG, Baekkeskov S. Glutamate decarboxylase and GABA in pancreatic islets: lessons from knock-out mice. Horm Metab Res 1999 May;31(5):340-4

(261)   Kubosaki A, Gross S, Miura J, Saeki K, Zhu M, Nakamura S, et al. Targeted disruption of the IA-2beta gene causes glucose intolerance and impairs insulin secretion but does not prevent the development of diabetes in NOD mice. diab 2004 Jul;53(7):1684-91.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15220191

(262)   Kubosaki A, Miura J, Notkins AL. IA-2 is not required for the development of diabetes in NOD mice. diabetol 2004 Jan;47(1):149-50.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=14614561

(263)   Kim SK, Tarbell KV, Sanna M, Vadeboncoeur M, Warganich T, Lee M, et al. Prevention of type I diabetes transfer by glutamic acid decarboxylase 65 peptide 206-220-specific T cells. Proc Natl Acad Sci U S A 2004 Sep 28;101(39):14204-9.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15381770

(264)   Burton AR, Vincent E, Arnold PY, Lennon GP, Smeltzer M, Li CS, et al. On the pathogenicity of autoantigen-specific T-cell receptors. Diabetes 2008 May;57(5):1321-30.http://www.ncbi.nlm.nih.gov/pubmed/18299317?ordinalpos=7&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(265)   Burton AR, Baquet Z, Eisenbarth GS, Tisch R, Smeyne R, Workman CJ, et al. Central Nervous System Destruction Mediated by Glutamic Acid Decarboxylase-Specific CD4+ T Cells. J Immunol 2010 Mar 26.PM:20348424

(266)   Zekzer D, Wong FS, Ayalon O, Millet I, Altieri M, Shintani S, et al. GAD-reactive CD4+ Th1 cells induce diabetes in NOD/SCID mice. J Clin Invest 1998 Jan 1;101(1):68-73.PM:9421467

(267)   Yoon JW, Yoon CS, Lim HW, Huang QQ, Kang Y, Pyun KH, et al. Control of autoimmune diabetes in NOD mice by GAD expression or suppression in beta cells. Science 1999 May 14;284(5417):1183-7

(268)   Gebe JA, Unrath KA, Yue BB, Miyake T, Falk BA, Nepom GT. Autoreactive human T-cell receptor initiates insulitis and impaired glucose tolerance in HLA DR4 transgenic mice. J Autoimmun 2008 Jun;30(4):197-206.http://www.ncbi.nlm.nih.gov/pubmed/17949947?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(269)   Quintana FJ, Carmi P, Cohen IR. DNA vaccination with heat shock protein 60 inhibits cyclophosphamide-accelerated diabetes. J Immunol 2002 Nov 15;169(10):6030-5.PM:12421990

(270)   Ablamunits V, Elias D, Reshef T, Cohen IR. Islet T cells secreting IFN-gamma in NOD mouse diabetes: arrest by p277 peptide treatment. J Autoimmun 1998 Feb;11(1):73-81

(271)   Elias D, Meilin A, Ablamunits V, Birk OS, Carmi P, Könen-Waisman S, et al. Hsp60 peptide therapy of NOD mouse diabetes induces a Th2 cytokine burst and downregulates autoimmunity to various b-cell antigens. diab 1997;46:758-64

(272)   Knip M, Karjalainen J, Akerblom HK. Islet cell antibodies are less predictive of IDDM among unaffected children in the general population than in sibs of children with diabetes. The Childhood Diabetes in Finland Study Group. Diab care 1998;21(10):1670-3

(273)   Bowman M, Atkinson MA. Heat shock protein therapy fails to prevent diabetes in NOD mice. diabetol 2002 Sep;45(9):1350-1.PM:12242470

(274)   Faideau B, Briand JP, Lotton C, Tardivel I, Halbout P, Jami J, et al. Expression of preproinsulin-2 gene shapes the immune response to preproinsulin in normal mice. J Immunol 2004 Jan 1;172(1):25-33.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=14688305

(275)   Devendra D, Paronen J, Moriyama H, Miao D, Eisenbarth GS, Liu E. Differential immune response to B:9-23 insulin 1 and insulin 2 peptides in animal models of type 1 diabetes. Journal of Autoimmunity 2004;23(1):17-26.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15236749

(276)   French MB, Allison J, Cram DS, Thomas HE, Dempsey-Collier M, Silva A, et al. Transgenic expression of mouse proinsulin II prevents diabetes in nonobese diabetic mice. diab 1996;46:34-9

(277)   Steptoe RJ, Ritchie JM, Harrison LC. Transfer of hematopoietic stem cells encoding autoantigen prevents autoimmune diabetes. J Clin Invest 2003 May;111(9):1357-63.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=12727927

(278)   DiLorenzo TP, Serreze DV. The good turned ugly: immunopathogenic basis for diabetogenic CD8+ T cells in NOD mice. Immunol Rev 2005 Apr;204:250-63.:250-63

(279)   Wenzlau JM, Juhl K, Yu L, Moua O, Sarkar SA, Gottlieb P, et al. The cation efflux transporter ZnT8 (Slc30A8) is a major autoantigen in human type 1 diabetes. Proc Natl Acad Sci U S A 2007 Oct 23;104(43):17040-5.http://www.ncbi.nlm.nih.gov/pubmed/17942684?ordinalpos=8&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(280)   Krishnamurthy B, Dudek NL, McKenzie MD, Purcell AW, Brooks AG, Gellert S, et al. Responses against islet antigens in NOD mice are prevented by tolerance to proinsulin but not IGRP. J Clin Invest 2006 Dec 1;116(12):3258-65.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=pubmed&cmd=Retrieve&dopt=AbstractPlus&list_uids=17143333&query_hl=7&itool=pubmed_docsum

(281)   Nakhooda AF, Like AA, Chappel CI, Murray FT, Marliss EB. The spontaneously diabetic Wistar rat: metabolic and morphologic studies. diab 2001;26:100-12

(282)   Like AA, Rossini AA. Spontaneous autoimmune diabetes mellitus in the BioBreeding/Worcester rat. Surv Synth Path Res 1984;3(2):131-8

(283)   Colle E, Guttmann RD, Seemayer T. Spontaneous diabetes mellitus syndrome in the rat. I. Association with the major histocompatibility complex. J Exp Med 1981;154(4):1237-42

(284)   Guttmann RD, Colle E, Michel F, Seemayer T. Spontaneous diabetes mellitus syndrome in the rat.  II. T lymphopenia and its association with clinical disease and pancreatic lymphocytic infiltration. J Immunol 1983;130(4):1732-5

(285)   Elder ME, Maclaren NK. Identification of profound peripheral T lymphocyte immunodeficiencies in the spontaneously diabetic BB rat. J Immunol 1983 Apr;130(4):1723-31.PM:6220066

(286)   Bellgrau D, Naji A, Silvers WK, Markmann JF, Barker CF. Spontaneous diabetes in BB rats: evidence for T cell dependent immune response defect. diabetol 1982;23(4):359-64

(287)   Woda BA, Like AA, Padden C, McFadden ML. Deficiency of phenotypic cytotoxic-suppressor T lymphocytes in the BB/W rat. J Immunol 1986 Feb 1;136(3):856-9.PM:2934480

(288)   Sellins KS, Gold DP, Bellgrau D. Resistance to tolerance induction in the diabetes-prone biobreeding rat as one manifestation of abnormal responses to superantigens. diabetol 1996;39(1):28-36

(289)   Crisa L, Mordes JP, Rossini AA. Autoimmune diabetes mellitus in the BB rat. Diabetes Metab Rev 1992;8(1):4-37

(290)   Mordes JP, Bortell R, Doukas J, Rigby M, Whalen B, Zipris D, et al. The BB/Wor rat and the balance hypothesis of autoimmunity. Diabetes Metab Rev 1996 Jul;12(2):103-9

(291)   Prins JB, Herberg L, den Bieman M, van Zutpen B. Genetic variation within and between lines of diabetes-prone and non-diabetes-prone BB rats: allele distribution of 8 protein markers. Lab Anim 2001;25:207-11

(292)   Butler L, Guberski DL, Like A. Changes in penetrance and onset of sontaneous diabetes in the BB/Wor rat. In: Shafrir E, editor. Frontier in Diabetes Research II. Lessons from Animal Diabetes III. 1990. p. 50-3.

(293)   Zipris D, Lien E, Nair A, Xie JX, Greiner DL, Mordes JP, et al. TLR9-signaling pathways are involved in Kilham rat virus-induced autoimmune diabetes in the biobreeding diabetes-resistant rat. J Immunol 2007 Jan 15;178(2):693-701.http://www.ncbi.nlm.nih.gov/sites/entrez?orig_db=PubMed&db=PubMed&cmd=Search&term=178%5Bvolume%5D%20AND%20693%5Bpage%5D%20AND%202007%5Bpdat%5D

(294)   Colle E, Fuks A, Poussier P, Edouard P, Guttman RD. Polygenic nature of spontaneous diabetes in the rat:  Permisive MHC haplotype and presence of the lymphopenic trait of the BB rat are not sufficient to produce susceptibility. diab 1992;41:1617-23

(295)   Jackson RA, Buse JB, Rifai R, Pelletier D, Milford EL, Carpenter CB, et al. Two genes required for diabetes in BB rats. Evidence from cyclical intercrosses and backcrosses. J Exp Med 1984;159(6):1629-36

(296)   Pandarpurkar M, Wilson-Fritch L, Corvera S, Markholst H, Hornum L, Greiner DL, et al. Ian4 is required for mitochondrial integrity and T cell survival. Proc Natl Acad Sci U S A 2003 Sep 2;100(18):10382-7.http://www.ncbi.nlm.nih.gov/pubmed/12930893?ordinalpos=6&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(297)   Lang JA, Kominski D, Bellgrau D, Scheinman RI. Partial activation precedes apoptotic death in T cells harboring an IAN gene mutation. Eur J Immunol 2004 Sep;34(9):2396-406.http://www.ncbi.nlm.nih.gov/pubmed/15307172?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(298)   Nitta T, Nasreen M, Seike T, Goji A, Ohigashi I, Miyazaki T, et al. IAN family critically regulates survival and development of T lymphocytes. PLoS Biol 2006 Apr;4(4):e103.http://www.ncbi.nlm.nih.gov/pubmed/16509771?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(299)   Ellerman KE, Like AA. Susceptibility to diabetes is widely distributed in normal class IIu haplotype rats. diabetol 2000 Jul;43(7):890-8.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=pubmed&cmd=Retrieve&dopt=AbstractPlus&list_uids=10952462&query_hl=25&itool=pubmed_docsum

(300)   Penhale WJ, Stumbles PA, Huxtable CR, Sutherland RJ, Pethick DW. Induction of diabetes in PVG/c strain rats by manipulation of the immune system. Autoimmunity 1990;7(2-3):169-79.PM:2104183

(301)   Martin AM, Maxson MN, Leif J, Mordes JP, Greiner DL, Blankenhorn EP. Diabetes-prone and diabetes-resistant BB rats share a common major diabetes susceptibility locus, iddm4: additional evidence for a "universal autoimmunity locus" on rat chromosome 4. diab 1999 Nov;48(11):2138-44.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=10535446

(302)   Kloting I, I, van den BJ, Kovacs P. Quantitative trait loci for blood glucose confirm diabetes predisposing and protective genes, Iddm4 and Iddm5r, in the spontaneously diabetic BB/OK rat. Int J Mol Med 1998 Nov;2(5):597-601.PM:9858659

(303)   Wallis RH, Wang K, Dabrowski D, Marandi L, Ning T, Hsieh E, et al. A novel susceptibility locus on rat chromosome 8 affects spontaneous but not experimentally induced type 1 diabetes. Diabetes 2007 Jun;56(6):1731-6.http://www.ncbi.nlm.nih.gov/pubmed/17389329?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(304)   Hornum L, Romer J, Markholst H. The diabetes-prone BB rat carries a frameshift mutation in Ian4, a positional candidate of Iddm1. diab 2002 Jun;51(6):1972-9.PM:12031988

(305)   MacMurray AJ, Moralejo DH, Kwitek AE, Rutledge EA, Van Yserloo B, Gohlke P, et al. Lymphopenia in the BB Rat Model of Type 1 Diabetes is Due to a Mutation in a Novel Immune-Associated Nucleotide (Ian)-Related Gene. Genome Res 2002 Jul;12(7):1029-39.PM:12097339

(306)   Hernandez-Hoyos G, Joseph S, Miller NG, Butcher GW. The lymphopenia mutation of the BB rat causes inappropriate apoptosis of mature thymocytes. Eur J Immunol 1999 Jun;29(6):1832-41.PM:10382745

(307)   Blankenhorn EP, Cort L, Greiner DL, Guberski DL, Mordes JP. Virus-induced autoimmune diabetes in the LEW.1WR1 rat requires Iddm14 and a genetic locus proximal to the major histocompatibility complex. diab 2009 Dec;58(12):2930-8.PM:19720792

(308)   Guberski DL, Thomas VA, Shek WR, Like AA, Handler ES, Rossini AA, et al. Induction of Type 1 diabetes by Kilham's rat virus in diabetes-resistant BB/Wor rats. Science 1991;254:1010-3.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=1658938

(309)   Sobel DO, Newsome J, Ewel CH, Bellanti JA, Abbassi V, Creswell K, et al. Poly I:C induces development of diabetes mellitus in BB rat. diab 1992;41(4):515-20

(310)   Like AA. Depletion of RT6.1+ lymphocytes alone is insufficient to induce diabetes in diabetes-resistant BB/WOR rats. Am J Pathol 1990;136:565-74

(311)   Ramanathan S, Bihoreau MT, Paterson AD, Marandi L, Gauguier D, Poussier P. Thymectomy and radiation-induced type 1 diabetes in nonlymphopenic BB rats. diab 2002 Oct;51(10):2975-81.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=12351436

(312)   Ellerman KE, Richards CA, Guberski DL, Shek WR, Like AA. Kilham rat virus triggers T-cell-dependent autoimmune diabetes in multiple strains of rat. diab 1996;45:557-62

(313)   Chung YH, Jun HS, Son M, Bao M, Bae HY, Kang Y, et al. Cellular and molecular mechanism for Kilham rat virus-induced autoimmune diabetes in DR-BB rats. J Immunol 2000 Sep 1;165(5):2866-76.PM:10946320

(314)   Zipris D, Lien E, Xie JX, Greiner DL, Mordes JP, Rossini AA. TLR activation synergizes with Kilham rat virus infection to induce diabetes in BBDR rats. J Immunol 2005 Jan 1;174(1):131-42.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15611235

(315)   Hornum L, Lundsgaard D, Markholst H. PolyI:C induction of diabetes is controlled by Iddm4 in rats with a full regulatory T cell pool. Ann N Y Acad Sci 2007 Sep;1110:65-72.:65-72

(316)   Blankenhorn EP, DeScipio C, Rodemich L, Cort L, Leif JH, Greiner DL, et al. Refinement of the Iddm4 diabetes susceptibility locus reveals TCRVbeta4 as a candidate gene. Ann N Y Acad Sci 2007 Apr;1103:128-31. Epub@2007 Mar 21.:128-31

(317)   Hornum L, DeScipio C, Markholst H, Troutman SA, Novak S, Leif J, et al. Comparative mapping of rat Iddm4 to segments on HSA7 and MMU6. Mamm Genome 2004 Jan;15(1):53-61.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=14727142

(318)   Kloting I, Schmidt S, Kovacs P. Mapping of novel genes predisposing or protecting diabetes development in the BB/OK rat. Biochem Biophys Res Commun 1998 Apr 17;245(2):483-6.PM:9571179

(319)   Jackerott M, Hornum L, Andreasen BE, Markholst H. Segregation of autoimmune type 1 diabetes in a cross between diabetic BB and brown Norway rats. J Autoimmun 1997 Feb;10(1):35-41.PM:9080298

(320)   Mordes JP, Cort L, Norowski E, Leif J, Fuller JM, Lernmark A, et al. Analysis of the rat Iddm14 diabetes susceptibility locus in multiple rat strains: identification of a susceptibility haplotype in the Tcrb-V locus. Mamm Genome 2009 Mar;20(3):162-9.PM:19205800

(321)   Jackson R, Rassi N, Crump A, Haynes BF, Eisenbarth GS. The BB diabetic rat. Profound T-cell lymphocytopenia. diab 1981;30(10):887-9

(322)   Ely JM, Greiner DL, Lubaroff DM, Fitch FW. Characterization of monoclonal antibodies that define rat T cell alloantigens. J Immunol 1983;130:2798-803

(323)   Greiner DL, Handler ES, Nakano K, Mordes JP, Rossini AA. Absence of the RT-6 T cell subset in diabetes-prone BB/W rats. J Immunol 1986;136:148-51

(324)   Groen H, Van Der Berk J, Nieuwenhuis P, Kampinga J. Peripheral T cells in diabetes prone (DP) BB rats are CD45R-negative. Thymus 1989;14:145-50

(325)   Spickett GP, Brandon MR, Mason DW, Williams AF, Woollett GR. MRC OX-22, a monoclonal antibody that labels a new subset of T lymphocytes and reacts with the high molecular weight form of the leukocyte-common antigen. J Exp Med 1983 Sep 1;158(3):795-810.PM:6224884

(326)   Thomas VA, Woda BA, Handler ES, Greiner DL, Mordes JP, Rossini AA. Altered expression of diabetes in BB/Wor rats by exposure to viral pathogens. diab 1991;40(2):255-8

(327)   Whalen BJ, Greiner DL, Mordes JP, Rossini AA. Adoptive transfer of autoimmune diabetes mellitus to athymic rats: synergy of CD4+ and CD8+ T cells and prevention by RT6+ T cells. J Autoimmun 1994 Dec;7(6):819-31.PM:7888038

(328)   Rossini AA, Mordes JP, Greiner DL, Nakano K, Appel MC, Handler ES. Spleen cell transfusion in the bio-breeding/worcester rat. J Clin Invest 1986;77:1399-401

(329)   Hillebrands JL, Whalen B, Visser JT, Koning J, Bishop KD, Leif J, et al. A regulatory CD4+ T cell subset in the BB rat model of autoimmune diabetes expresses neither CD25 nor Foxp3. J Immunol 2006 Dec 1;177(11):7820-32

(330)   Holm TL, Lundsgaard D, Markholst H. Characteristics of rat CD4(+)CD25(+) T cells and their ability to prevent not only diabetes but also insulitis in an adoptive transfer model in BB rats. Scand J Immunol 2006 Jul;64(1):17-29

(331)   Georgiou HM, Lagarde AC, Bellgrau D. T cell dysfunction in the diabetes-prone BB rat: a role for thymic migrants that are not T cell precursors. J Exp Med 1988;167:132-48

(332)   Bellgrau D, Redd JM, Sellins KS. Peculiar T-cell signaling does not preclude positive selection in the diabetes-prone BB rat. diab 1994;43(1):47-52

(333)   Hosszufalusi N, Chan E, Granger G, Charles MA. Quantitative analyses comparing all major spleen cell phenotypes in BB and normal rats: autoimmune imbalance and double negative T cells associated with resistant, prone and diabetic animals. J Autoimmun 1992 Jun;5(3):305-18.PM:1388637

(334)   Moore JK, Bellgrau D. Promiscuous activation and cell cycle entry in T cells from autoimmune animals. Transplant Proc 1999 May;31(3):1606-10.PM:10331020

(335)   Bieg S, Möller C, Olsson T, Lernmark Å. The lymphopenia (lyp) gene controls the intrathymic cytokine ratio in congenic BioBreeding rats. diabetol 1997;40:786-92

(336)   Bieg S. Differential expression of p95vav in primary lymphoid tissue of BB rats congenic for the lymphopenia gene. Autoimmunity 1999;30(1):37-42.PM:10433093

(337)   Zadeh HH, Greiner DL, Wu DY, Tausche F, Goldschneider I. Abnormalities in the export and fate of recent thymic emigrants in diabetes-prone BB/W rats. Autoimmunity 1996;24(1):35-46.PM:8937686

(338)   Moore JK, Scheinman RI, Bellgrau D. The identification of a novel T cell activation state controlled by a diabetogenic gene. J Immunol 2001 Jan 1;166(1):241-8.PM:11123298

(339)   Ramanathan S, Norwich K, Poussier P. Antigen activation rescues recent thymic emigrants from programmed cell death in the BB rat. J Immunol 1998 Jun 15;160(12):5757-64.PM:9637485

(340)   Delemarre FG, Simons PJ, de Heer HJ, Drexhage HA. Signs of immaturity of splenic dendritic cells from the autoimmune prone biobreeding rat: consequences for the in vitro expansion of regulator and effector T cells. J Immunol 1999 Feb 1;162(3):1795-801.PM:9973444

(341)   Delemarre FG, Hoogeveen PG, Haan-Meulman M, Simons PJ, Drexhage HA. Homotypic cluster formation of dendritic cells, a close correlate of their state of maturation. Defects in the biobreeding diabetes-prone rat. J Leukoc Biol 2001 Mar;69(3):373-80.PM:11261783

(342)   Iwakoshi NN, Greiner DL, Rossini AA, Mordes JP. Diabetes prone BB rats are severely deficient in natural killer T cells. Autoimmunity 1999;31(1):1-14.PM:10593564

(343)   Tullin S, Farris P, Petersen JS, Hornum L, Jackerott M, Markholst H. A pronounced thymic B cell deficiency in the spontaneously diabetic BB rat. J Immunol 1997 Jun 1;158(11):5554-9.PM:9164980

(344)   Rozing J, Coolen C, Tielen FJ, Weegenaar J, Schuurman HJ, Greiner DL, et al. Defects in the thymic epithelial stroma of diabetes prone BB rats. Thymus 1989;14(1-3):125-35.PM:2623736

(345)   Bellgrau D, Lagarde A-C. Cytotoxic T-cell precursors with low-level CD8 in the diabetes-prone biobreeding rat: implications for the generation of an autoimmune T-cell repertoire. Proc Natl Acad Sci USA 1990;87:313-7

(346)   Groen H, Klatter FA, Brons NH, Mesander G, Nieuwenhuis P, Kampinga J. Abnormal thymocyte subset distribution and differential reduction of CD4+ and CD8+ T cell subsets during peripheral maturation in diabetes-prone BioBreeding rats. J Immunol 1996 Feb 1;156(3):1269-75.PM:8558007

(347)   Plamondon C, Kottis V, Brideau C, Metroz-Dayer MD, Poussier P. Abnormal thymocyte maturation in spontaneously diabetic BB rats involves the deletion of CD4-8+ cells. J Immunol 1990 Feb 1;144(3):923-8.PM:1688592

(348)   Whalen BJ, Weiser P, Marounek J, Rossini AA, Mordes JP, Greiner DL. Recapitulation of normal and abnormal BioBreeding rat T cell development in adult thymus organ culture. J Immunol 1999 Apr 1;162(7):4003-12.PM:10201921

(349)   McKeever U, Mordes JP, Greiner DL, Appel MC, Rozing J, Handler ES, et al. Adoptive transfer of autoimmune diabetes and thyroiditis to athymic rats. Proc Natl Acad Sci U S A 1990 Oct;87(19):7618-22.PM:2217193

(350)   Sarkar P, Crisa L, McKeever U, Bortell R, Handler E, Mordes JP, et al. Loss of RT6 message and most circulating T cells after thymectomy of diabetes prone BB rats. Autoimmunity 1994;18(1):15-22.PM:7999952

(351)   Jung CG, Kamiyama T, Agui T. Elevated apoptosis of peripheral T lymphocytes in diabetic BB rats. Immunology 1999 Dec;98(4):590-4.PM:10594693

(352)   Crispe IN, Dao T, Klugewitz K, Mehal WZ, Metz DP. The liver as a site of T-cell apoptosis: graveyard, or killing field? Immunol Rev 2000 Apr;174:47-62.PM:10807506

(353)   Scott J. The spontaneously diabetic BB rat: sites of the defects leading to autoimmunity and diabetes mellitus. A review. Curr Top Microbiol Immunol 1990;156:1-14.PM:2199161

(354)   Francfort JW, Naji A, Silvers WK, Barker CF. The influence of T-lymphocyte precursor cells and thymus grafts on the cellular immunodeficiencies of the BB rat. diab 1985;34:1134-8

(355)   Like A, Rossini AA, Guberski DL, Appel MC. Spontaneous diabetes mellitus: Reversal and prevention in the BB/W rat with antiserum to rat lymphocytes. Science 1979;206:1421-3

(356)   Like AA, Biron CA, Weringer EJ, Byman K, Sroczynski E, Guberski DL. Prevention of diabetes in BioBreeding/Worcester rats with monoclonal antibodies that recognize T lymphocytes or natural killer cells. J Exp Med 1986;164(4):1145-59

(357)   Barlow AK, Like AA. Anti-CD2 monoclonal antibodies prevent spontaneous and adoptive transfer of diabetes in the BB/Wor rat. Am J Pathol 1992 Nov;141(5):1043-51.PM:1359788

(358)   Like AA, Kislauskis E, Williams RR, Rossini AA. Neonatal thymectomy prevents spontaneous diabetes mellitus in the BB/W rat. Science 1982;216(4546):644-6

(359)   Murase N, Lieberman I, Nalesnik M, Mintz D, Todo S, Drash AL, et al. Prevention of spontaneous diabetes in BB rats with FK 506. Lancet 1990 Aug 11;336(8711):373-4.PM:1697397

(360)   Nicoletti F, Meroni PL, Barcellini W, Grasso S, Borghi MO, Lunetta M, et al. FK-506 prevents diabetes in diabetes-prone BB/Wor rats. Int J Immunopharmacol 1991;13(7):1027-30.PM:1722192

(361)   Sadelain MW, Qin HY, Sumoski W, Parfrey N, Singh B, Rabinovitch A. Prevention of diabetes in the BB rat by early immunotherapy using Freund's adjuvant. J Autoimmun 1990 Dec;3(6):671-80

(362)   Like AA, Guberski DL, Butler L. Influence of environmental viral agents on frequency and tempo of diabetes mellitus in BB/Wor rats. diab 1991;40:259-62

(363)   Dryberg T, Schwimmbeck PL, Oldstone MBA. Inhibition of diabetes in BB rats by virus infection. J Clin Invest 1988;81:928-31

(364)   Greiner DL, Malkani S, Kanaitsuka T, Bortell R, Doukas J, Rigby M, et al. The T cell marker RT6 in a rat model of autoimmune diabetes. Adv Exp Med Biol 1997;419:209-16.PM:9193656

(365)   Rossini AA, Faustman D, Woda BA, Like AA, Szymanski I, Mordes JP. Lymphocyte transfusions prevent diabetes in the Bio-Breeding/ Worcester rat. J Clin Invest 1984;74(1):39-46

(366)   Burstein D, Mordes JP, Greiner DL, Stein D, Nakamura N, Handler ES, et al. Prevention of diabetes in BB/Wor rat by single transfusion of spleen cells. Parameters that affect degree of protection. diab 1989 Jan;38(1):24-30.PM:2642432

(367)   Ramanathan S, Poussier P. T cell reconstitution of BB/W rats after the initiation of insulitis precipitates the onset of diabetes. J Immunol 1999 May 1;162(9):5134-42.PM:10227984

(368)   Satoh J, Seino H, Shintani S, Tanaka S, Ohteki T, Masuda T, et al. Inhibition of type 1 diabetes in BB rats with recombinant human tumor necrosis factor-alpha. J Immunol 1990 Sep 1;145(5):1395-9.PM:2384663

(369)   Whalen BJ, Marounek J, Weiser P, Appel MC, Greiner DL, Mordes JP, et al. BB rat thymocytes cultured in the presence of islets lose their ability to transfer autoimmune diabetes. diab 2001 May;50(5):972-9.http://www.ncbi.nlm.nih.gov/sites/entrez?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=11334440

(370)   Brugman S, Klatter FA, Visser JT, Wildeboer-Veloo AC, Harmsen HJ, Rozing J, et al. Antibiotic treatment partially protects against type 1 diabetes in the Bio-Breeding diabetes-prone rat. Is the gut flora involved in the development of type 1 diabetes? Diabetologia 2006 Sep;49(9):2105-8

(371)   Kawano K, Hirashima T, Moris S, Saitoh Y, Kurosumi M, Natori T. New inbred strain of Long-Evans Tokushima lean rats with IDDM without lymphopenia. diab 1991;40(11):1375-81

(372)   Yokoi N. Identification of a major gene responsible for type 1 diabetes in the Komeda diabetes-prone rat. Exp Anim 2005 Apr;54(2):111-5.http://www.ncbi.nlm.nih.gov/pubmed/15897618?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(373)   Yokoi N, Kanazawa M, Kitada K, Tanaka A, Kanazawa Y, Suda S, et al. A non-MHC locus essential for autoimmune type I diabetes in the Komeda diabetes-prone rat. J Clin Invest 1997;100:2015-21

(374)   Yokoi N, Hayashi C, Fujiwara Y, Wang HY, Seino S. Genetic reconstitution of autoimmune type 1 diabetes with two major susceptibility genes in the rat. Diabetes 2007 Feb;56(2):506-12.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=pubmed&cmd=Retrieve&dopt=AbstractPlus&list_uids=17259398&query_hl=5&itool=pubmed_docsum

(375)   Komeda K, Noda M, Terao K, Kuzuya N, Kanazawa M, Kanazawa Y. Establishment of two substrains, diabetes-prone and non-diabetic, from Long-Evans Tokushima Lean (LETL) rats. Endocr J 1998 Dec;45(6):737-44.PM:10395228

(376)   Yokoi N, Komeda K, Wang HY, Yano H, Kitada K, Saitoh Y, et al. Cblb is a major susceptibility gene for rat type 1 diabetes mellitus. Nat Genet 2002 Aug;31(4):391-4.PM:12118252

(377)   Kosoy R, Yokoi N, Seino S, Concannon P. Polymorphic variation in the CBLB gene in human type 1 diabetes. Genes Immun 2004 May;5(3):232-5.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=14961073

(378)   Payne F, Smyth DJ, Pask R, Barratt BJ, Cooper JD, Twells RC, et al. Haplotype tag single nucleotide polymorphism analysis of the human orthologues of the rat type 1 diabetes genes Ian4 (Lyp/Iddm1) and Cblb. diab 2004 Feb;53(2):505-9.PM:14747305

(379)   Bergholdt R, Taxvig C, Eising S, Nerup J, Pociot F. CBLB variants in type 1 diabetes and their genetic interaction with CTLA4. J Leukoc Biol 2005 Apr;77(4):579-85

(380)   Lenzen S, Tiedge M, Elsner M, Lortz S, Weiss H, Jorns A, et al. The LEW.1AR1/Ztm-iddm rat: a new model of spontaneous insulin-dependent diabetes mellitus. diabetol 2001 Sep;44(9):1189-96.PM:11596676

(381)   Jorns A, Kubat B, Tiedge M, Wedekind D, Hedrich HJ, Kloppel G, et al. Pathology of the pancreas and other organs in the diabetic LEW.1AR1/Ztm- iddm rat, a new model of spontaneous insulin-dependent diabetes mellitus. Virchows Arch 2004 Feb;444(2):183-9.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=14735361

(382)   Arndt T, Wedekind D, Weiss H, Tiedge M, Lenzen S, Hedrich HJ, et al. Prevention of spontaneous immune-mediated diabetes development in the LEW.1AR1-iddm rat by selective CD8+ T cell transfer is associated with a cytokine shift in the pancreas-draining lymph nodes. diabetol 2009 Jul;52(7):1381-90.PM:19367386

(383)   Jorns A, Gunther A, Hedrich HJ, Wedekind D, Tiedge M, Lenzen S. Immune cell infiltration, cytokine expression, and beta-cell apoptosis during the development of type 1 diabetes in the spontaneously diabetic LEW.1AR1/Ztm-iddm rat. Diabetes 2005 Jul;54(7):2041-52

(384)   Lipes MA, Rosenzweig A, Tan KN, Tanigawa G, Ladd D, Seidman JG, et al. Progression to diabetes in nonobese diabetic (NOD) mice with transgenic T cell receptors. Science 1993;259(5098):1165-9.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=8267690

(385)   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

(386)   Verdaguer J, Yoon J-W, Anderson B, Averill N, Utsugi T, Park B-J, et al. Acceleration of spontaneous diabetes in TCR-b-transgenic nonobese diabetic mice by b-cell cytotoxic CD8+ T cells expressing identical endognous TCR-a chains. J Immunol 1996;157:4726-35

(387)   Verdaguer J, Schmidt D, Amrani A, Anderson B, Averill N, Santamaria P. Spontaneous autoimmune diabetes in monoclonal T cell nonobese diabetic mice. J Exp Med 1997;186:1663-76

(388)   Graser RT, DiLorenzo TP, Wang F, Christianson GJ, Chapman HD, Roopenian DC, et al. Identification of a CD8 T cell that can independently mediate autoimmune diabetes development in the complete absence of CD4 T cell helper functions. J Immunol 2000 Apr 1;164(7):3913-8.PM:10725754

(389)   Wong FS, Visintin I, Wen L, Flavell RA, Janeway CA. CD8 T cell clones from young nonobese diabetic (NOD) islets can transfer rapid onset of diabetes in NOD mice in the absence of CD4 cells. J Exp Med 1996;183:67-76

(390)   Wegmann DR, Shehadeh N, Lafferty KJ, Norbury-Glaser N, Gill RG, Daniel D. Establishment of islet-specific T cell lines and clones from islet isografts placed in spontaneously diabetic NOD mice. J Autoimmun 1993;6:517-27

(391)   Yang Y, Santamaria P. Dissecting autoimmune diabetes through genetic manipulation of non-obese diabetic mice. diabetol 2003 Nov;46(11):1447-64.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=14586501

(392)   Haskins K, Wegmann D. Diabetogenic T-cell clones. diab 1996;45:1299-305

(393)   Chaparro RJ, Burton AR, Serreze DV, Vignali DA, DiLorenzo TP. Rapid identification of MHC class I-restricted antigens relevant to autoimmune diabetes using retrogenic T cells. J Immunol Methods 2008 Jun 1;335(1-2):106-15.PM:18439618

(394)   Holst J, Vignali KM, Burton AR, Vignali DA. Rapid analysis of T-cell selection in vivo using T cell-receptor retrogenic mice. Nat Methods 2006 Mar;3(3):191-7.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=pubmed&cmd=Retrieve&dopt=AbstractPlus&list_uids=16489336&query_hl=7&itool=pubmed_docsum

(395)   Holst J, Szymczak-Workman AL, Vignali KM, Burton AR, Workman CJ, Vignali DA. Generation of T-cell receptor retrogenic mice. Nat Protoc 2006;1(1):406-17

(396)   Arnold PY, Burton AR, Vignali DA. Diabetes incidence is unaltered in glutamate decarboxylase 65-specific TCR retrogenic nonobese diabetic mice: generation by retroviral-mediated stem cell gene transfer. J Immunol 2004 Sep 1;173(5):3103-11.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15322170

(397)   Zhang L, Jasinski JM, Kobayashi M, Davenport B, Johnson K, Davidson H, et al. Analysis of T cell receptor beta chains that combine with dominant conserved TRAV5D-4*04 anti-insulin B:9-23 alpha chains. J Autoimmun 2009 Mar 13.PM:19286348

(398)   Katz JD, Wang B, Haskins K, Benoist C, Mathis D. Following a diabetogenic T cell from genesis through pathogenesis. Cell 1993;74:1089-100

(399)   Shi FD, Flodstrom M, Balasa B, Kim SH, Van Gunst K, Strominger JL, et al. Germ line deletion of the CD1 locus exacerbates diabetes in the NOD mouse. Proc Natl Acad Sci U S A 2001 Jun 5;98(12):6777-82.PM:11390999

(400)   Masteller EL, Warner MR, Ferlin W, Judkowski V, Wilson D, Glaichenhaus N, et al. Peptide-MHC class II dimers as therapeutics to modulate antigen-specific T cell responses in autoimmune diabetes. J Immunol 2003 Nov 15;171(10):5587-95.http://www.ncbi.nlm.nih.gov/pubmed/14607967?ordinalpos=4&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(401)   Jasinski JM, Yu L, Nakayama M, Li MM, Lipes MA, Eisenbarth GS, et al. Transgenic insulin (B:9-23) T-cell receptor mice develop autoimmune diabetes dependent upon RAG genotype, H-2g7 homozygosity, and insulin 2 gene knockout. Diabetes 2006 Jul;55(7):1978-84.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=pubmed&cmd=Retrieve&dopt=AbstractPlus&list_uids=16804066&query_hl=9&itool=pubmed_docsum

(402)   Kanagawa O, Shimizu J, Vaupel BA. Thymic and postthymic regulation of diabetogenic CD8 T cell development in TCR transgenic nonobese diabetic (NOD) mice. J Immunol 2000 May 15;164(10):5466-73.PM:10799914

(403)   Miller JF, Morahan G, Allison J, Hoffmann M. A transgenic approach to the study of peripheral T-cell tolerance. Immunol Rev 1991 Aug;122:103-16.PM:1937538

(404)   Oldstone MBA, Nerenberg M, Southern P, Price J, Lewicki H. Virus infection triggers insulin-dependent diabetes mellitus in a transgenic model: Role of anti-self (virus) immune response. Cell 1991;65:319-31

(405)   Ohashi PS, Oehen S, Buerki K, Pircher H, Ohashi CT, Odermatt B, et al. Ablation of "tolerance" and induction of diabetes by virus infection in viral antigen transgenic mice. Cell 1991;65:305-17

(406)   Apostolou I, von Boehmer H. The TCR-HA, INS-HA transgenic model of autoimmune diabetes: limitations and expectations. J Autoimmun 2004 Mar;22(2):111-4.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=14987738

(407)   von Herrath MG, Coon B, Lewicki H, Mazarguil H, Gairin JE, Oldstone MB. In vivo treatment with a MHC class I-restricted blocking peptide can prevent virus-induced autoimmune diabetes. J Immunol 1998 Nov 1;161(9):5087-96.PM:9794447

(408)   Laufer TM, von Herrath MG, Grusby MJ, Oldstone MBA, Glimcher LH. Autoimmune diabetes can be induced in transgenic major histocompatibility complex class II-deficient mice. J Exp Med 1993;178:589-96

(409)   von Herrath MG, Dockter J, Oldstone MBA. How virus induces a rapid or slow onset insulin-dependent diabetes mellitus in a transgenic model. Immunity 1994;1:231-42

(410)   Lo D, Freedman J, Hesse S, Palmiter RD, Brinster RL, Sherman LA. Peripheral tolerance to an islet cell-specific hemagglutinin transgene affects both CD4+ and CD8+ T cells. Eur J Immunol 1992 Apr;22(4):1013-22.PM:1348026

(411)   Harlan DM, Hengartner H, Huang ML, Kang YH, Abe R, Moreadith RW, et al. Mice expressing both B7-1 and viral glycoprotein on pancreatic beta cells along with glycoprotein-specific transgenic T cells develop diabetes due to a breakdown of T-lymphocyte unresponsiveness. Proc Natl Acad Sci U S A 1994 Apr 12;91(8):3137-41.PM:7512724

(412)   King C, Mueller HR, Malo CM, Murali-Krishna K, Ahmed R, King E, et al. Interleukin-4 acts at the locus of the antigen-presenting dendritic cell to counter-regulate cytotoxic CD8+ T-cell responses. Nat Med 2001 Feb;7(2):206-14.PM:11175852

(413)   Apostolou I, Sarukhan A, Klein L, von Boehmer H. Origin of regulatory T cells with known specificity for antigen. Nat Immunol 2002 Aug;3(8):756-63.PM:12089509

(414)   Morgan DJ, Liblau R, Scott B, Fleck S, McDevitt HO, Sarvetnick N, et al. CD8(+) T cell-mediated spontaneous diabetes in neonatal mice. J Immunol 1996;157:978-83

(415)   Vizler C, Bercovici N, Cornet A, Cambouris C, Liblau RS. Role of autoreactive CD8+ T cells in organ-specific autoimmune diseases: insight from transgenic mouse models. Immunol Rev 1999 Jun;169:81-92.PM:10450510

(416)   Degermann S, Reilly C, Scott B, Ogata L, von Boehmer H, Lo D. On the various manifestations of spontaneous autoimmune diabetes in rodent models. Eur J Immunol 1994 Dec;24(12):3155-60.PM:7528672

(417)   Scott B, Liblau R, Degermann S, Marconi LA, Ogata L, Caton AJ, et al. A role for non-MHC genetic polymorphism in susceptibility to spontaneous autoimmunity. Immunity 1994 Apr;1(1):73-83.PM:7889402

(418)   Kurts C, Heath WR, Kosaka H, Miller JF, Carbone FR. The peripheral deletion of autoreactive CD8+ T cells induced by cross-presentation of self-antigens involves signaling through CD95 (Fas, Apo-1). J Exp Med 1998 Jul 20;188(2):415-20.PM:9670055

(419)   Blanas E, Carbone FR, Allison J, Miller JF, Heath WR. Induction of autoimmune diabetes by oral administration of autoantigen. Science 1996;274(5293):1707-9

(420)   Heath WR, Allison J, Hoffmann MW, Schonrich G, Hammerling G, Arnold B, et al. Autoimmune diabetes as a consequence of locally produced interleukin-2. Nature 1992 Oct 8;359(6395):547-9.PM:1406974

(421)   Lesage S, Hartley S, Wilson JD, Goodnow CC. T cell tolerance in a transgenic mouse model for autoimmune diabetes. Keystone Symposia Regulation of Immunity and Autoimmunity , 115. 2001.

Ref Type: Abstract

(422)   Wong FS, Dittel BN, Janeway CAJ. Transgenes and knockout mutations in animal models of type 1 diabetes and multiple sclerosis. Immunol Rev 1999 Jun;169:93-104

(423)   Elias D, Markovits D, Reshef T, 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

(424)   Renold AE, Soeldner JS, Steinke J. Immunological studies with homologous and heterologous pancreatic insulin in the cow. Ciba Foundation Colloquia on Endocrinology 15, 122-134. 1964. 1964.

Ref Type: Journal (Full)

(425)   Renold AE, Steinke J, Soeldner JS, Smith RS, Antoniades HN. Response of calves and heifers to the prolonged administration of porcine and bovine insulin. XVIIth Scientific Meeting of the Protein Foundation; 1965 p. 121-2.

(426)   LeCompte PM, Steinke J, Soeldner JS, Renold AE. Changes in the islets of Langerhans in cows injected with heterologous and homologous insulin. diab 1966;15(8):586-96

(427)   Renold AE, Steinke J, Soeldner JS, Antomiades HN, Smith RM. Immunological response to the prolonged administration of heterologous and homologous insulin in cattle. J Clin Invest 1966;45(5):702-13

(428)   Grodsky GM, Feldman R, Toreson WE, Lee JC. Diabetes mellitus in rabbits immunized with insulin. diab 1966;15:579-85

(429)   Kloppel G, Freytag G. Insulin antibodies and immune insulitis in rabbits immunized with bovine or porcine insulin components. Horm Metab Res 1975;7:25-30

(430)   Glimcher LH, Schroer JA, Chan C, Shevach EM. Fine specificity of cloned insulin-specific T cell hybridomas: evidence supporting a role for tertiary conformation. J Immunol 1983 Dec;131(6):2868-74.PM:6196405

(431)   Kahn CR, Rosenthal AS. Immunologic reactions to insulin:  Insulin allergy, insulin resistance, and the autimmune insulin syndrome. Diab care 1979;2:283-95

(432)   Abiru N, Maniatis AK, Yu L, Miao D, Moriyama H, Wegmann D, et al. Peptide and MHC specific breaking of humoral tolerance to native insulin with the B:9-23 peptide in diabetes prone and normal mice. diab 2001 Jun;50:1274-81.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=11375327

(433)   Moriyama H, Wen L, Abiru N, Liu E, Yu L, Miao D, et al. Induction and acceleration of insulitis/diabetes in mice with a viral mimic (polyinosinic-polycytidylic acid) and an insulin self-peptide. Proc Natl Acad Sci U S A 2002 Apr 16;99(8):5539-44

(434)   Liu E, Abiru N, Moriyama H, Miao D, Eisenbarth GS. Induction of Insulin Autoantibodies and Protection from Diabetes with Subcutaneous Insulin B:9-23 Peptide Without Adjuvant. Annals New York Academy of Sciences 2002;958:224-7

(435)   Wen L, Peng J, Li Z, Wong FS. The effect of innate immunity on autoimmune diabetes and the expression of toll-like receptors on pancreatic islets. J Immunol 2004 Mar 1;172(5):3173-80.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=14978124

(436)   Liu E, Moriyama H, Abiru N, Paronen J, Devendra D, Finkelman FD, et al. Preventing peptide-induced anaphylaxis: addition of C-terminal amino acids to produce a neutral isoelectric point. J Allergy Clin Immunol 2004 Sep;114(3):607-13.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15356565

(437)   Liu E, Moriyama H, Abiru N., Miao D, Yu L, Taylor RM, et al. Anti-peptide autoantibodies and fatal anaphylaxis in NOD mice in response to insulin self-peptides B:9-23 and B:13-23. J Clin Invest 2002;110(7):1021-7

(438)   Devendra D, Jasinski J, Melanitou E, Nakayama M, Li M, Hensley B, et al. Interferon-{alpha} as a Mediator of Polyinosinic:Polycytidylic Acid-Induced Type 1 Diabetes. diab 2005 Sep;54(9):2549-56.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=16123342

(439)   Graham KL, Sanders N, Tan Y, Allison J, Kay TW, Coulson BS. Rotavirus infection accelerates type 1 diabetes in mice with established insulitis. J Virol 2008 Jul;82(13):6139-49

(440)   Paronen J, Liu E, Moriyama H, Devendra D, Ide A, Taylor R, et al. Genetic differentiation of Poly I : C from B : 9-23 peptide induced experimental autoimmune diabetes. Journal of Autoimmunity 2004;22(4):307-13.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15120754

(441)   von Herrath MG, Guerder S, Lewicki H, Flavell RA, Oldstone MB. Coexpression of B7-1 and viral ("self") transgenes in pancreatic beta cells can break peripheral ignorance and lead to spontaneous autoimmune diabetes. Immunity 1995 Dec;3(6):727-38.PM:8777718

(442)   Pechhold K, Karges W, Blum C, Boehm BO, Harlan DM. Beta cell-specific CD80 (B7-1) expression disrupts tissue protection from autoantigen-specific CTL-mediated diabetes. J Autoimmun 2003 Feb;20(1):1-13.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=12604308

(443)   Wen L, Chen NY, Tang J, Sherwin R, Wong FS. The regulatory role of DR4 in a spontaneous diabetes DQ8 transgenic model. J Clin Invest 2001 Apr;107(7):871-80.PM:11285306

(444)   Allison J, Stephens LA, Kay TW, Kurts C, Heath WR, Miller JF, et al. The threshold for autoimmune T cell killing is influenced by B7-1. Eur J Immunol 1998 Mar;28(3):949-60.PM:9541590

(445)   Herrera PL, Harlan DM, Fossati L, Izui S, Huarte J, Orci L, et al. A CD8+ T-lymphocyte-mediated and CD4+ T-lymphocyte-independent autoimmune diabetes of early onset in transgenic mice. diabetol 1994 Dec;37(12):1277-9.PM:7534735

(446)   Subudhi SK, Zhou P, Yerian LM, Chin RK, Lo JC, Anders RA, et al. Local expression of B7-H1 promotes organ-specific autoimmunity and transplant rejection. J Clin Invest 2004 Mar;113(5):694-700.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=14991067

(447)   Serreze DV, Marron MP, DiLorenzo TP. "Humanized" HLA transgenic NOD mice to identify pancreatic beta cell autoantigens of potential clinical relevance to type 1 diabetes. Ann N Y Acad Sci 2007 Apr;1103:103-11. Epub@2007 Mar 21.:103-11

(448)   Macchiarini F, Manz MG, Palucka AK, Shultz LD. Humanized mice: are we there yet? J Exp Med 2005 Nov 21;202(10):1307-11.PM:16301740

(449)   Fugger L. Human autoimmunity genes in mice. Curr Opin Immunol 2000 Dec;12(6):698-703.PM:11102775

(450)   Herman AE, Tisch RM, Patel SD, Parry SL, Olson J, Noble JA, et al. Determination of glutamic acid decarboxylase 65 peptides presented by the type I diabetes-associated HLA-DQ8 class II molecule identifies an immunogenic peptide motif. J Immunol 1999 Dec 1;163(11):6275-82

(451)   Liu J, Purdy LE, Rabinovitch S, Jevnikar AM, Elliott JF. Major DQ8-restricted T-cell epitopes for human GAD65 mapped using human CD4, DQA1*0301, DQB1*0302 transgenic IA(null) NOD mice. diab 1999 Mar;48(3):469-77

(452)   Havari E, Lennon-Dumenil AM, Klein L, Neely D, Taylor JA, McInerney MF, et al. Expression of the B7.1 costimulatory molecule on pancreatic beta cells abrogates the requirement for CD4 T cells in the development of type 1 diabetes. J Immunol 2004 Jul 15;173(2):787-96.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15240665

(453)   Gebe JA, Unrath KA, Falk BA, Ito K, Wen L, Daniels TL, et al. Age-dependent loss of tolerance to an immunodominant epitope of glutamic acid decarboxylase in diabetic-prone RIP-B7/DR4 mice. Clin Immunol 2006 Dec;121(3):294-304.http://www.ncbi.nlm.nih.gov/pubmed/16979383?ordinalpos=3&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum

(454)   Marietta E, Black K, Camilleri M, Krause P, Rogers RS, III, David C, et al. A new model for dermatitis herpetiformis that uses HLA-DQ8 transgenic NOD mice. J Clin Invest 2004 Oct;114(8):1090-7.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15489956

(455)   Taneja V, Griffiths M, Behrens M, Luthra HS, David CS. Auricular chondritis in NOD.DQ8.Abetao (Ag7-/-) transgenic mice resembles human relapsing polychondritis. J Clin Invest 2003 Dec;112(12):1843-50.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=14679179

(456)   Marron MP, Graser RT, Chapman HD, Serreze DV. Functional evidence for the mediation of diabetogenic T cell responses by HLA-A2.1 MHC class I molecules through transgenic expression in NOD mice. Proceedings of the National Academy of Sciences of the United States of America 2002;99(21):13753-8

(457)   Banuelos SJ, Shultz LD, Greiner DL, Burzenski LM, Gott B, Lyons BL, et al. Rejection of human islets and human HLA-A2.1 transgenic mouse islets by alloreactive human lymphocytes in immunodeficient NOD-scid and NOD-Rag1(null)Prf1(null) mice. Clin Immunol 2004 Sep;112(3):273-83.http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list_uids=15308121

(458)   Jarchum I, Takaki T, DiLorenzo TP. Efficient culture of CD8(+) T cells from the islets of NOD mice and their use for the study of autoreactive specificities. J Immunol Methods 2008 Nov 30;339(1):66-73.PM:18782577

(459)   Jarchum I, Baker JC, Yamada T, Takaki T, Marron MP, Serreze DV, et al. In vivo cytotoxicity of insulin-specific CD8+ T-cells in HLA-A*0201 transgenic NOD mice. diab 2007 Oct;56(10):2551-60.PM:17620420

(460)   Shultz LD, Lyons BL, Burzenski LM, Gott B, Chen X, Chaleff S, et al. Human lymphoid and myeloid cell development in NOD/LtSz-scid IL2R gamma null mice engrafted with mobilized human hemopoietic stem cells. J Immunol 2005 May 15;174(10):6477-89.http://www.ncbi.nlm.nih.gov/pubmed/17332083?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_RVDocSum1

(461)   Brehm MA, Bortell R, Diiorio P, Leif J, Laning J, Cuthbert A, et al. Human immune system development and rejection of human islet allografts in spontaneously diabetic NOD-Rag1null IL2rgammanull Ins2Akita mice. diab 2010 Sep;59(9):2265-70.PM:20570944