当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 糖尿病学杂志 > 2005年 > 第7期 > 正文
编号:11256637
NOD B-cells Are Insufficient to Incite T-Cell-Mediated Anti-islet Autoimmunity
     1 Department of Pediatrics, Vanderbilt Children’s Hospital, Vanderbilt University, Nashville, Tennessee

    2 Harrison Department of Surgical Research, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania

    ABSTRACT

    Although it is well established that B-cells are required for the development of diabetes in the nonobese diabetic (NOD) mouse, the nature of their role remains unknown. Herein, we investigate the hypothesis that B-cells in this autoimmune background actively disrupt the tolerant state of those T-cells with which they interact. We demonstrate that NOD B-cells express elevated levels of crucial molecules involved in antigen presentation (including CD21/35, major histocompatibility complex class II, and CD40), alterations that invite the possibility of inappropriate T-cell activation. However, when chimeric animals are generated in which all B-cells are NOD-derived, a tolerant state is maintained. These data demonstrate that although B-cells are required for the development of autoimmunity, they are not sufficient to disrupt established tolerance. Moreover, non-B-cell antigen-presenting cells may be the critical actors in the establishment of the tolerant state; this function may be absent in NOD mice as they are characterized by deficient professional antigen-presenting cell function.

    Autoimmune diabetes results from the destruction of insulin secreting -cells in the pancreas by autoreactive T-cells. Although T-cells are the end-stage effectors in the NOD model, a significant line of evidence points to crucial dysfunctions in the antigen-presenting cell (APC) compartments (macrophages, dendritic cells, B-cells) as the reason that peripheral tolerance fails to be maintained (1eC7). In particular, B-cells with their ability to process and present antigen are required for the development of diabetes (8eC12).

    However, the mechanism by which B-cells promote the development of diabetes remains unknown. Data from our laboratory suggests that the requisite function of B-cells results from the inability of non--cell APC subsets to promote proliferation of T-cells in the nonobese diabetic (NOD) mouse (13,14). Therefore, the NOD B-cell pool is the only efficient coactivator of T-cell reactivity. Yet, it remains unknown whether peripheral tolerance is disrupted by interaction with these B-cells or rather is never established secondary to inactivity on the part of other thymic or peripheral APCs. Here, we address this question with a chimeric system in which all B-cells are NOD derived.

    Progeny resulting from intercrosses between the NOD mouse and other nonautoimmune strains are uniformly protected from the development of diabetes (15eC17). However, lethal irradiation of these mice and reconstitution with NOD bone marrow restores the progression to islet destruction and disease. The addition of F1-derived bone marrow to this inoculum has been reported to restore the protection from diabetes. These data suggest that cells derived from the F1 bone marrow prevent the realization of the pathogenic potential inherent in NOD bone marrow derivatives (18,19). The adaptation of this system presented here permits the evaluation of T-cell activation and tolerance in the presence of NOD-derived B-cells. These data suggest that tolerance can be maintained in the presence of this repertoire, that autoimmunity likely results from the absence of critical tolerance promoting factors within another APC subset, and that NOD-derived B-cells are insufficient to drive T-cell-mediated autoimmunity.

    RESEARCH DESIGN AND METHODS

    NOD/LtJ, NOD.NONThy1.1, C57BL/6, and C57BL/6 e蘉TeC/eC mice were purchased from The Jackson Laboratories (Bar Harbor, ME). All other strains (F1.g7, F1e蘉TeC/eC, NOD e蘉TeC/eC, and B6.g7) were derived from stocks maintained in our facility. All mice were housed under specific pathogen-free barrier conditions and maintained according to the guidelines for use and care of laboratory animals as set forth by the University of Pennsylvania. All NOD mice and chimera were monitored weekly for the development of diabetes by blood glucose measurement with Accu-Check Advantage test strips (Boehringer Mannheim, Indianapolis, IN). Two consecutive daily glucose measurements >250 mg/dl constituted a diagnosis of diabetes. Control groups were compared with experimental at the end point of each diabetes incidence experiment with the 2 test.

    Generation of chimeric mice.

    Chimeric mice were produced as described previously by Matthieu et al. (19). To generate mixed chimera, F1 recipients received sublethal irradiation (1,000 rad) in a divided dose (500 x 2) on consecutive days. For full NOD chimera, lethal irradiation (1,200 rad) was employed in a divided dose (600 x 2) on consecutive days. Immediately after the second irradiation treatment, mice received 20 million nondepleted bone marrow cells from the appropriate donor. Mixed inocula were prepared as a 1:1 ratio with 10 million of each donor type. All recipient mice were female to maximize the penetrance of diabetes within these groups.

    Flow cytometry.

    One million splenocytes were suspended in biotin-free RPMI containing 0.1% azide and 3% FCS and surface stained in 96-well plates with the appropriate monoclonal antibody: RA3eC6B2 biotin (anti-CD45R/B220), RM4eC5 allophycocyanin (anti-CD4), 10eC3.6 PE (anti-I-Ag7), OX-7 PE (anti-CD90.1/Thy-1.1), 30-H12 biotin (anti-CD90.2/Thy-1.2) (BD PharMingen). Biotin-conjugated monoclonal antibodies were subsequently stained with streptavidin-RED670 (Life Technologies); cells were washed no fewer than two times before the addition of the secondary reagent. All samples were analyzed on a FACSCalibur flow cytometer (Becton Dickinson, Mountain View, CA) using CellQuest software.

    CFSE labeling of lymphocytes.

    Splenocytes were labeled with 5-(and eC6)-carboxyfluorescein diacetate succinimidyl ester (CFSE) (Molecular Probes, Eugene, OR) as previously described (20). Briefly, splenocytes were resuspended at a concentration of 10 x 106 cells/ml in serum-free IMDM (Gibco/BRL, Gaithersburg, MD) at 37°C. An equal volume of a 1:350 dilution of the CFSE stock (5 mmol/l in DMSO) in 37°C serum-free IMDM was then added to the cell preparation, which was subsequently incubated for 5 min at 37°C. CFSE labeling was quenched by adding an equal volume of heat-inactivated FCS, whereupon cells were washed twice and resuspended in IMDM containing 10% heat-inactivated FCS.

    In vitro T-cell stimulations.

    CFSE-labeled cells as described in the previous section were plated in 24-well plates at a density of 1 x 106 total cells in 1 ml of media containing 10% heat-inactivated FCS with the designated amount of anti-CD3 (145eC2C11) and 4 e/ml anti-CD28 (37.51) antibodies. All cells were incubated for 65eC70 h at 37°C in 5% CO2. After incubation, the cultured cells were harvested and stained with allophycocyanin-conjugated anti-CD4 (RM4eC5) to allow the identification of CFSE-labeled CD4+ T-cells using FACSCalibur (Becton Dickinson, Mountain View, CA). In all, 10,000 CD4+ events were collected and division was tracked utilizing a live cell gate that included the blasting cells, as determined by forward and side scatter. Division history of CD4+ T-cells was analyzed as previously described, based on the property of CFSE-labeled cells to lose half of their fluorescence intensity with each round of division (21).

    Probability calculation.

    The data generated by CFSE-labeled cultures was analyzed to determine the potential for activation inherent in the precursor pool. To perform this calculation, we determined the number of activated precursors required to produce all the daughter cells in a given division peak (P), a factor we term "precursor equivalents," by using the following formula:

    where P is the peak number.

    The probability of generating a daughter cell in peak P is given by dividing the number of precursor equivalents for that division by the total number of equivalents for all divided peaks (P > 1).

    RESULTS

    Upregulation of antigen-presentation machinery on NOD B-cells.

    Previous data from our laboratory has indicated that the B-cell is the only efficient coactivator of T-cells within the NOD system (13,14). Therefore, characteristics of B-cell APCs determine the nature of T-cell activation. Given the crucial role of this cell class in diabetogenesis, we undertook a careful analysis of the cell surface expression of molecules involved in antigen presentation on the NOD B-cell.

    NOD B-cell function was assessed with respect to antigen presentation by comparing surface levels of antigen presenting and costimulatory molecules to the levels observed on the control B6.g7 strain (Fig. 1), which is congenic to NOD at the major histocompatibility complex (MHC) locus. Class II, CD40, CD19, and CD21/35 were upregulated on splenic NOD B-cells; we have previously reported on the role of the complement receptor complex in the development of diabetes (22). Levels of CD80, CD86, CD54, and CD1 were indistinguishable between these strains (data not shown). Together, these data paint a picture of a B-cell that has extended its ability to acquire antigen (complement receptor), to present those antigens (Class II), to respond to stimulation (CD19), and to activate those T-cells which it might engage (CD40). Analysis of this increased expression in splenocytes from T-cell deficient (CeC/eC) NOD mice revealed a similar increase in surface levels, indicating that these alterations did not result from interaction with autoreactive T-cells (data not shown).

    F1e蘉TeC/eC chimera: Diabetes in the presence of NOD B-cells

    The upregulation of several key mediators of antigen presentation invited the possibility that NOD B-cells may inappropriately activate NOD T-cells. Moreover, as B-cells are the only competent APCs within the lymph node microenvironment, alteration in B-cell functional status may have profound consequences for the maintenance of peripheral tolerance (13). We hypothesized that the activated nature of NOD B-cells may permit them to actively disrupt the tolerant state. To test this hypothesis, we constructed chimera in which all B-cells were NOD derived. Bone marrow with this reconstituting profile was obtained from F1 e蘉TeC/eC mice which were produced from crosses between NOD e蘉TeC/eC and B6 e蘉TeC/eC B-cell deficient parents. Mice reconstituted with mixtures of F1 e蘉TeC/eC and NOD bone marrow demonstrated an absence of F1 B-cells in the periphery despite the presence of NOD- derived B-cells (Fig. 2). In addition, fluorescence-activated cell sorter analysis to track the MHC class I disparities indicated that the T-cell compartment was evenly divided among the donor strains in the mixed chimera by 15 weeks after reconstitution (Fig. 2). No differences were detected in the percentages of CD4 cells, CD8 cells, B-cells, or macrophage/dendritic cells between the reconstituted groups; analysis of absolute splenocyte numbers at the termination of the experiment produced expected numbers of cells (>100 x 106 per spleen) and no significant difference between the groups (data not shown). Although the B-cell activity of these mice was derived wholly from the autoimmune-prone NOD strain, these mice were protected from spontaneous diabetes for the >70-week duration of the study (Fig. 3). Further analysis of splenocytes from these chimera revealed the same alterations in class II, CD21/35, CD19, and CD40 present in the NOD strain (data not shown). These data indicate that NOD-derived B-cells do not incite autoimmunity and suggest that another APC subset likely participated in the establishment of a tolerant state.

    "Normal" MHC molecules are not required for the protection of F1 chimera.

    The data from F1e蘉TeC/eC chimera indicate that diabetes resistance can be maintained even in the absence of F1 B-cells; this finding suggests that the function of bone-marrow-derived F1 professional APCs may provide the protective quality we have observed. However, the mechanism of action of these cells remained unknown. It was also plausible that the presence of MHC molecules distinct from I-Ag7 leads to the selection of a peripheral lymphocyte repertoire devoid of islet reactivity. This explanation is supported by studies in which the presence of alternative MHC molecules in the NOD mouse prevents diabetes (23eC26). Nonetheless, there is also ample evidence of pathogenic specificities that are not deleted in these hybrids (27,28). Alternatively, the presence of normal F1-derived professional APCs could lead to T-cell tolerance following contact with peripheral autoantigens by dint of other tolerizing functions. We sought to exclude the first possibility as the primary cause of protection seen from F1 bone marrow. To analyze this contention, we created NODxB6.g7 progeny, a substrain we have designated F1.g7. This strain expresses only NOD-derived MHC molecules and hence could be more permissive to diabetes induction than F1s derived from the B6 strain in which the MHC will be heterozygous (g7 and b).

    Chimera were generated with the irradiation protocols described above. NOD bone marrow was collected from NOD.Thy1.1 donors to permit tracking of T-cell reconstitution as these F1.g7 mice are MHC congenic to the NOD. Mixed chimeras reconstituted with an average of 31% NOD T-cells; T-cells in full chimera were more than 95% NOD-derived (Fig. 4); no differences were detected in overall proportion of B-cells or CD8 cells (data not shown). As shown in Fig. 5, full NOD chimera became diabetic around 25 weeks after reconstitution. Mixed NOD/F1.g7 mice were completely protected from diabetes (P < 0.05) despite the presence of significant numbers of NOD-derived T-lymphocytes. These data indicate that the presence of non-NOD MHC alleles is not solely responsible for the protection from diabetes seen previously but rather that some other function possibly within the professional APC subclass serves to establish or maintain peripheral tolerance.

    Cellular proliferation in F1.g7 mice: F1 phenotype does not prevent diabetes.

    Previous data from our laboratory and others has demonstrated a hypoproliferative T-cell phenotype in the NOD strain, which develops around 4eC6 weeks of age and precedes the onset of diabetes (13,14,29,30). As these chimeric mice were protected from diabetes, we sought to analyze their proliferative response to determine whether NOD proliferation characteristics tracked with the development of diabetes. Both the proliferative distribution of splenocytes at maximal stimulation and the dose response physiology were determined. The parameters studied in these assays did not serve to distinguish these groups in any way. As shown in Fig. 6, splenocytes from both protected and susceptible mice at maximal stimulation demonstrated a level of proliferation intermediate between that seen for NOD and B6 CD4 T-cells. These cultures generated some daughter progeny in the latter cell divisions, a feature that demonstrated an improvement over NOD stimulation levels but did not reach that seen in B6 cultures. In addition, the dose-response curves of both groups were not distinguishable from each other and were identical to that seen in the NOD strain (Fig. 7). This analysis indicates that dose response and proliferative phenotypes do not predict progression to disease. The possibility remains that these protected chimera may retain susceptibility to some of the autoimmune syndromes that can be induced in NOD mice (31eC37). This aspect will require further investigation.

    DISCUSSION

    Overall, these data indicate that the presence of normal non-B-cell APCs may prevent the development of autoimmunity. We have further demonstrated that the protective function of these "normal" bone marrow derivatives as characterized by previous investigators does not rely on the expression of normal MHC molecules but rather can still be exerted even when these cells also expressed the diabetes associated MHC antigen, I-Ag7. Although our prior data had indicated that NOD B-cells were primarily responsible for the activation of diabetogenic T-cell specificities, these autoimmune strain B-cells were not able to promote diabetes in the presence of other normal APCs (9,13,14,38). Rather, only in the case of the NOD mouse (where B-cells provide the majority of coactivating stimuli in the presence of an effete professional APC compartment) does diabetes ensue.

    The mechanism by which these professional APC compartments promote stable tolerance remains to be elucidated. We and others have previously suggested that tolerance induction in the NOD mouse may be impeded by the hypoproliferative phenotype illustrated in the CD4 T-cell compartment (4,14,39,40). However, the data here suggest that the proliferative phenotype of the peripheral T-cell compartment is not predictive of disease development. That both diabetic and resistant F1 chimera were indistinguishable by all estimates of their proliferative capacity suggests that at least part of the tolerance-inducing mechanism operates independently of proliferative achievement. Whether these chimera are stably tolerant or can be induced to develop other aspects of autoimmunity will be an important future assessment of their immunologic status (32eC37,41,42). The presence of normal APCs may hinder the targeting of islet tissue while leaving open the possibility that latent tendencies to autoimmunity are unrealized in the absence of other environmental triggers.

    Prior studies have amply demonstrated the requirement of B-cell APC function for diabetogenesis and T-cell activation in NOD mice but have not indicated the mechanism by which B-cells participate in the development of autoimmunity. We have considered whether NOD-derived B-cells may actively disrupt the tolerant state and our study has negated the hypothesis that autoimmune diabetes results directly from interaction with B-cells from the NOD background (8eC12,38). Our initial analysis of the B-cell compartment indicated a number of anomalies in cell surface molecule expression and painted a picture of heightened costimulatory activity which might promote autoreactivity. Nonetheless, when F1 chimera were constructed in which all B-cells were NOD derived, diabetes did not result despite the maintenance of these features within the B-cell compartment. These data confirm the critical role of other APCs in establishing or maintaining peripheral tolerance. In addition, B-cell-deficient NOD mice may be protected from the development of diabetes by the ineffectual properties of the remaining professional APC compartments rather than by virtue of being tolerant to autoantigens (13). The data presented here may also have important implications for understanding clinical disease entities and for designing preventative therapies. There has been a report of the development of diabetes within a B-cell-deficient kindred (43). Although prior data in murine systems suggests that autoreactive T-cells would not be activated in the absence of B-cells, the data here suggest that the B-cell-deficient NOD may not be tolerant to autoantigens (8,11eC13,38,42,44). Moreover, the clinical data may suggest that the T-cell-activating and tolerizing capacity of professional APCs are separable traits. Therefore, it may be possible to restore the T-cell-activating capacity of professional APCs within the NOD system and thereby permit the development of diabetes in the absence of B-cells. Secondarily, these data also suggest that although B-cell depletion may prevent the progression of diabetes, it may not induce a stable state of tolerance without additional manipulations aimed at restoring the function of the professional APC compartment. In this regard, clinical trials that consider B-cell depleting therapies (such as rituximab for the prevention of diabetes) should examine manifestations of tolerance during prolonged follow-up.

    Intriguingly, deficiencies in APC activation and function are emerging as critical factors in other models of autoimmunity. The recent characterization of the CARD15/NOD2 gene in inflammatory bowel disease suggests that mutations in the promoter of this toll-like gene product limit macrophage function in those human populations most susceptible to Crohn’s disease (45eC49). The inability to produce optimal inflammatory signaling may hinder T-cell targeting and trafficking and result in an increase in the likelihood of inappropriate T-cell responses.

    That the presence of a small population of surrogate APCs derived from a nonautoimmune strain can halt the development of diabetes is an exciting observation that may be extended to yet other models of autoimmunity. These data suggest that the deficiencies within the professional antigen-presenting compartments prevent the establishment or maintenance of tolerance and suggest that tolerance induction requires contact with these professional APC lineages. Future studies should determine the activity of these cells and whether their mechanism of action impinges directly on the T-cell compartment or possibly modifies the function of the B-cell pool.

    ACKNOWLEDGMENTS

    This research was supported by Grants DK54215 and DK49814 from the National Institutes of Health. D.J.M. is supported by the American Diabetes Association Physician Scientist Training Grant.

    FOOTNOTES

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

    APC, antigen-presenting cell; CFSE, carboxyfluorescein diacetate succinimidyl ester; IMDM, Iscove’s modification of Dulbecco’s medium; MHC, major histocompatibility complex

    REFERENCES

    Charre S, Rosmalen JG, Pelegri C, Alves V, Leenen PJ, Drexhage HA, Homo-Delarche F: Abnormalities in dendritic cell and macrophage accumulation in the pancreas of nonobese diabetic (NOD) mice during the early neonatal period. Histol Histopathol17 :393 eC401,2002

    Alleva DG, Johnson EB, Wilson J, Beller DI, Conlon PJ: SJL and NOD macrophages are uniquely characterized by genetically programmed, elevated expression of the IL-12(p40) gene, suggesting a conserved pathway for the induction of organ-specific autoimmunity. J Leukoc Biol69 :440 eC448,2001

    Rosmalen JG, Homo-Delarche F, Durant S, Kap M, Leenen PJ, Drexhage HA: Islet abnormalities associated with an early influx of dendritic cells and macrophages in NOD and NODscid mice. Lab Invest80 :769 eC777,2000

    Piganelli JD, Martin T, Haskins K: Splenic macrophages from the NOD mouse are defective in the ability to present antigen. Diabetes47 :1212 eC1218,1998

    Serreze DV: Autoimmune diabetes results from genetic defects manifest by antigen presenting cells. FASEB J7 :1092 eC1096,1993

    Lo D, Reilly CR, Scott B, Liblau R, McDevitt HO, Burkly LC: Antigen-presenting cells in adoptively transferred and spontaneous autoimmune diabetes. Eur J Immunol23 :1693 eC1698,1993

    Serreze DV, Gaskins HR, Leiter EH: Defects in the differentiation and function of antigen presenting cells in NOD/Lt mice. J Immunol150 :2534 eC2543,1993

    Serreze DV, Chapman HD, Varnum DS, Hanson MS, Reifsnyder PC, Richard SD, Fleming SA, Leiter EH, Shultz LD: B-lymphocytes are essential for the initiation of T cell-mediated autoimmune diabetes: analysis of a new "speed congenic" stock of NOD.Ig mu null mice. J Exp Med184 :2049 eC2053,1996

    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. Diabetes46 :941 eC946,1997

    Chiu PP, Serreze DV, Danska JS: Development and function of diabetogenic T-cells in B-cell-deficient nonobese diabetic mice. Diabetes50 :763 eC770,2001

    Akashi T, Nagafuchi S, Anzai K, Kondo S, Kitamura D, Wakana S, Ono J, Kikuchi M, Niho Y, Watanabe T: Direct evidence for the contribution of B cells to the progression of insulitis and the development of diabetes in non-obese diabetic mice. Int Immunol9 :1159 eC1164,1997

    Yang M, Charlton B, Gautam AM: Development of insulitis and diabetes in B cell-deficient NOD mice. J Autoimmun10 :257 eC260,1997

    Greeley SA, Moore DJ, Noorchashm H, Noto LE, Rostami SY, Schlachterman A, Song HK, Koeberlein B, Barker CF, Naji A: Impaired activation of islet-reactive CD4 T cells in pancreatic lymph nodes of B cell-deficient nonobese diabetic mice. J Immunol167 :4351 eC4357,2001

    Noorchashm H, Moore DJ, Noto LE, Noorchashm N, Reed AJ, Reed AL, Song HK, Mozaffari R, Jevnikar AM, Barker CF, Naji A: Impaired CD4 T cell activation due to reliance upon B cell-mediated costimulation in nonobese diabetic (NOD) mice. J Immunol165 :4685 eC4696,2000

    Bendelac A, Boitard C, Bach JF, Carnaud C: Neonatal induction of allogeneic tolerance prevents T cell-mediated autoimmunity in NOD mice. Eur J Immunol19 :611 eC616,1989

    Wicker LS, Miller BJ, Chai A, Terada M, Mullen Y: Expression of genetically determined diabetes and insulitis in the nonobese diabetic (NOD) mouse at the level of bone marrow-derived cells. Transfer of diabetes and insulitis to nondiabetic (NOD X B10) F1 mice with bone marrow cells from NOD mice. J Exp Med167 :1801 eC1810,1988

    Serreze DV, Leiter EH, Worthen SM, Shultz LD: NOD marrow stem cells adoptively transfer diabetes to resistant (NOD x NON)F1 mice. Diabetes37 :252 eC255,1988

    Mathieu C, Casteels K, Bouillon R, Waer M: Protection against autoimmune diabetes in mixed bone marrow chimeras: mechanisms involved. J Immunol158 :1453 eC1457,1997

    Mathieu C, Bouillon R, Rutgeerts O, Waer M: Induction of mixed bone marrow chimerism as potential therapy for autoimmune (type I) diabetes: experience in the NOD model. Transplant Proc27 :640 eC641,1995

    Lyons AB, Parish CR: Determination of lymphocyte division by flow cytometry. J Immunol Methods171 :131 eC137,1994

    Wells AD, Gudmundsdottir H, Turka LA: Following the fate of individual T cells throughout activation and clonal expansion. Signals from T cell receptor and CD28 differentially regulate the induction and duration of a proliferative response. J Clin Invest100 :3173 eC3183,1997

    Noorchashm H, Moore DJ, Lieu YK, Noorchashm N, Schlachterman A, Song HK, Barker CF, Naji A: Contribution of the innate immune system to autoimmune diabetes: a role for the CR1/CR2 complement receptors. Cell Immunol195 :75 eC79,1999

    McDevitt H: The role of MHC class II molecules in the pathogenesis and prevention of Type I diabetes. Adv Exp Med Biol490 :59 eC66,2001

    Singer SM, Tisch R, Yang XD, Sytwu HK, Liblau R, McDevitt HO: Prevention of diabetes in NOD mice by a mutated I-Ab transgene. Diabetes47 :1570 eC1577,1998

    Hanson MS, Cetkovic-Cvrlje M, Ramiya VK, Atkinson MA, Maclaren NK, Singh B, Elliott JF, Serreze DV, Leiter EH: Quantitative thresholds of MHC class II I-E expressed on hemopoietically derived antigen-presenting cells in transgenic NOD/Lt mice determine level of diabetes resistance and indicate mechanism of protection. J Immunol157 :1279 eC1287,1996

    Parish NM, Chandler P, Quartey-Papafio R, Simpson E, Cooke A: The effect of bone marrow and thymus chimerism between non-obese diabetic (NOD) and NOD-E transgenic mice, on the expression and prevention of diabetes. Eur J Immunol23 :2667 eC2675,1993

    Parish NM, Acha-Orbea H, Simpson E, Qin SX, Lund T, Cooke A: A comparative study of T-cell receptor V beta usage in non-obese diabetic (NOD) and I-E transgenic NOD mice. Immunology78 :606 eC610,1993

    Bohme J, Schuhbaur B, Kanagawa O, Benoist C, Mathis D: MHC-linked protection from diabetes dissociated from clonal deletion of T cells. Science249 :293 eC295,1990

    Zipris D, Crow AR, Delovitch TL: Altered thymic and peripheral T-lymphocyte repertoire preceding onset of diabetes in NOD mice. Diabetes40 :429 eC435,1991

    Salojin KV, Zhang J, Madrenas J, Delovitch TL: T-cell anergy and altered T-cell receptor signaling: effects on autoimmune disease. Immunol Today19 :468 eC473,1998

    Baker D, Rosenwasser OA, O’Neill JK, Turk JL: Genetic analysis of experimental allergic encephalomyelitis in mice. J Immunol155 :4046 eC4051,1995

    Bernard NF, Ertug F, Margolese H: High incidence of thyroiditis and anti-thyroid autoantibodies in NOD mice. Diabetes41 :40 eC46,1992

    Humphreys-Beher MG: Animal models for autoimmune disease-associated xerostomia and xerophthalmia. Adv Dent Res10 :73 eC75,1996

    Krause I, Tomer Y, Elias D, Blank M, Gilburd B, Cohen IR, Shoenfeld Y: Inhibition of diabetes in NOD mice by idiotypic induction of SLE. J Autoimmun13 :49 eC55,1999

    Many MC, Maniratunga S, Varis I, Dardenne M, Drexhage HA, Denef JF: Two-step development of Hashimoto-like thyroiditis in genetically autoimmune prone non-obese diabetic mice: effects of iodine-induced cell necrosis. J Endocrinol147 :311 eC320,1995

    Silveira PA, Baxter AG: The NOD mouse as a model of SLE. Autoimmunity34 :53 eC64,2001

    Baxter AG, Healey D, Cooke A: Mycobacteria precipitate autoimmune rheumatic disease in NOD mice via an adjuvant-like activity. Scand J Immunol39 :602 eC606,1994

    Noorchashm H, Lieu YK, Noorchashm N, Rostami SY, Greeley SA, Schlachterman A, Song HK, Noto LE, Jevnikar AM, Barker CF, Naji A: I-Ag7-mediated antigen presentation by B-cells is critical in overcoming a checkpoint in T cell tolerance to islet beta cells of nonobese diabetic mice. J Immunol163 :743 eC750,1999

    Bergerot I, Arreaza G, Cameron M, Chou H, Delovitch TL: Role of T-cell anergy and suppression in susceptibility to IDDM. Res Immunol148 :348 eC358,1997

    Salojin K, Zhang J, Cameron M, Gill B, Arreaza G, Ochi A, Delovitch TL: Impaired plasma membrane targeting of Grb2-murine son of sevenless (mSOS) complex and differential activation of the Fyn-T cell receptor (TCR)-zeta-Cbl pathway mediate T cell hyporesponsiveness in autoimmune nonobese diabetic mice. J Exp Med186 :887 eC897,1997

    Becker KG: Comparative genetics of type 1 diabetes and autoimmune disease: common loci, common pathways Diabetes48 :1353 eC1358,1999

    Braley-Mullen H, Yu S: Early requirement for B cells for development of spontaneous autoimmune thyroiditis in NOD.H-2h4 mice. J Immunol165 :7262 eC7269,2000

    Martin S, Wolf-Eichbaum D, Duinkerken G, Scherbaum WA, Kolb H, Noordzij JG, Roep BO: Development of type 1 diabetes despite severe hereditary B-lymphocyte deficiency. N Engl J Med345 :1036 eC1040,2001

    Serreze DV, Fleming SA, Chapman HD, Richard SD, Leiter EH, Tisch RM: B-cells are critical antigen-presenting cells for the initiation of T cell-mediated autoimmune diabetes in nonobese diabetic mice. J Immunol161 :3912 eC3918,1998

    Ahmad T, Armuzzi A, Bunce M, Mulcahy-Hawes K, Marshall SE, Orchard TR, Crawshaw J, Large O, de Silva A, Cook JT, Barnardo M, Cullen S, Welsh KI, Jewell DP: The molecular classification of the clinical manifestations of Crohn’s disease. Gastroenterology122 :854 eC866,2002

    Bonen DK, Ogura Y, Nicolae DL, Inohara N, Saab L, Tanabe T, Chen FF, Foster SJ, Duerr RH, Brant SR, Cho JH, Nunez G: Crohn’s disease-associated NOD2 variants share a signaling defect in response to lipopolysaccharide and peptidoglycan. Gastroenterology124 :140 eC146,2003

    Gutierrez O, Pipaon C, Inohara N, Fontalba A, Ogura Y, Prosper F, Nunez G, Fernandez-Luna JL: Induction of Nod2 in myelomonocytic and intestinal epithelial cells via nuclear factor-kappa B activation. J Biol Chem277 :41701 eC41705,2002

    McGovern DP, van Heel DA, Ahmad T, Jewell DP: NOD2 (CARD15), the first susceptibility gene for Crohn’s disease. Gut49 :752 eC754,2001

    Murillo L, Crusius JB, van Bodegraven AA, Alizadeh BZ, Pena AS: CARD15 gene and the classification of Crohn’s disease. Immunogenetics54 :59 eC61,2002(Daniel J. Moore, Hooman N)