Abstract
Nearly 70% of newly produced B cells express autoreactive antigen receptors and must be silenced to prevent autoimmunity. Failure of silencing mechanisms is apparent in type 1 diabetes (T1D), where islet antigen-specific B cells appear critical for development of disease. Evidence for a B cell role in T1D includes success of B cell targeted anti-CD20 therapy, which delays T1D progression in both NOD mice and new onset patients. Demonstrating the importance of specificity, NOD mice whose B cell repertoire is biased toward insulin reactivity show increased disease development, while bias away from insulin reactivity largely prevents disease. Finally, though not required for illness, high affinity insulin autoantibodies are often the first harbingers of T1D. B cell cytokine production and auto-antigen presentation to self-reactive T cells are likely important in pathogenesis. Here we review B cell function, as described above, in T1D in humans and the non-obese diabetic (NOD) mouse. We will discuss recent broad-based B cell depletion studies and how they may provide the basis for refinement of future treatments for the disorder.
Keywords: Type 1 diabetes (T1D), B lymphocytes, Autoimmunity
Introduction
Type 1 diabetes (T1D) has traditionally been considered a T cell-mediated autoimmune disorder. It is certainly true that the pathogenic destruction of insulin-secreting pancreatic beta cells occurs via direct interaction with autoreactive T cells. However, an earlier breech in B cell tolerance or ignorance is also a major contributor to disease. This conclusion is based in part on the success of anti-CD20 (Rituximab) therapy, which by broadly depleting B cells delays disease progression, preserving beta cell function in both non-obese diabetic (NOD) mice and new onset human patients [1–3]. In T1D, B cells have been shown to promote autoimmunity through a number of mechanisms including, but not limited to: autoantibody production [4,5], antigen presentation [6–9], and the secretion of pro-inflammatory cytokines [10,11].
Here, we review the extant literature related to B cell function in T1D in humans and the non-obese diabetic (NOD) mouse. We discuss what can be learned from the broad based B cell depletion studies that have been completed in mouse and man, and speculate as to how these findings may refine future curative and preventative strategies for the disorder.
2. The paradox of B cell requirements for development of T1D
The importance of B cells in the spontaneous development of autoimmune diabetes (T1D) in NOD mice was clearly established using NOD. Igµ null mice that lack B cells [12]. These mice are resistant to T1D, but disease develops after reconstitution of a polyclonal B cell compartment [13]. Similarly, B cell depletion by chronic treatment with anti-IgM antibodies protects from disease development [14]. Further indicative of a requirement for B cell specificity are findings that biasing the B cell repertoire with an immunoglobulin heavy chain transgene that raises the frequency of insulin-specific B cells to a few percent greatly increases the penetrance and decreases the time of onset of disease [15]. Repertoire bias away from insulin reactivity largely prevents disease development. Thus islet antigen specificity appears key to B cell participation in T1D. It is somewhat paradoxical that in some cases modest disease is seen in the absence of B cells, and islet antigen-specific T cells can transfer disease to NOD. scid recipients lacking B cells [16,17]. Why is this? Development of disease following transfer of TCR transgenic T cells may simply reflect the ability of nonphysiologically high numbers of islet antigen-specific T cells with pathogenic potential to compensate for inefficient antigen presentation resulting from the absence of B cells. Furthermore, lymphopenic conditions in scid that promote rapid homeostatic expansion of transferred autoreactive T cells may also contribute to disease development.
In humans, this paradox is seen in the oft-cited study of a T1D patient that was deficient in B cells due to a Bruton’s tyrosine kinase (Btk) mutation causing X-linked agammaglobulinemia (XLA) [18]. It is noteworthy that the particular Btk mutation seen in this patient usually does not result in complete loss of B cells [19]. Keep in mind, as noted above, that B cell depleting therapy has efficacy in human T1D [3]. Thus, on balance, evidence indicates the B cells are important in development of T1D in both mouse and man.
3. B cells intrinsic defects in NOD contribute to breech in tolerance
At least three mechanisms: clonal deletion, receptor editing, and anergy normally function to eliminate and/or inactivate potentially dangerous auto-reactive B cells. Among these, available evidence suggests anergy is the most prominent silencing mechanism [20]. Anergic B cells survive in the periphery for a brief period of time, yet, despite continued availability of unoccupied antigen receptors (BCR), they are unresponsive to antigen stimulation [21,22]. The anergic state is labile, as it is dependent on continuous occupancy of antigen receptors and their transduction of inhibitory signals [23]. It is important to note that B cells bearing BCR with high autoantigen affinity can also persist in a naïve state in the periphery if they recognize only low avidity antigens. These cells are termed “ignorant” because despite binding autoantigen, BCR signaling does not occur due to lack of receptor aggregation. Since most ambient insulin is monomeric it seems likely that under normal circumstances insulin-specific B cells should be naive by virtue of ignorance unless they have the ability to recognize insulin bound to its receptor. Nonetheless, there is evidence that in T1D-resistant mouse backgrounds many insulin-specific B cells are eliminated by receptor editing or clonal deletion, while others are anergic (Hinman and Cambier, manuscript in preparation).
B cell participation in T1D in NOD may result from a break in tolerance or ignorance. Supporting the possibility that anergy is faulty in NOD mice are studies that explore the integrity of MD4 anti-HEL tolerance. In the NOD genetic background, MD4 anti-hen egg lysozyme (HEL) B cells undergo efficient clonal deletion upon encounter with membrane bound HEL. Deletion is comparable to that seen in autoimmunity resistant C57BL/6 mice. In mice expressing soluble HEL antigen, anti-HEL B cells are normally silenced primarily by anergy. However, in the NOD background the anergic status of HEL-specific B cells appears unstable [24]. This is true whether soluble antigen is expressed systemically, or limited to the pancreas by use of the insulin promoter [25]. We have explored the status of insulin specific B cells in NOD, and found that a large proportion of these cells become activated prior to onset of diabetes, and present antigen to CD4 T cells (Hinman and Cambier, manuscript in preparation). Thus B cell intrinsic mechanisms that normally function to confer and maintain B cell anergy are defective in NOD mice.
At least twenty-five genetic loci contribute to T1D in NOD. Cox et al. have mapped aspects of defective B cell anergy to loci on chromosome 1 (Idd5) and 4 (Idd9/11) [25]. In addition to faulty B cell tolerance, NOD mice have enlarged populations of marginal zone B cells [26]. This trait is shared by other models of autoimmunity, most notably lupus in (NZB × NZW)F1 [27]. Marginal zone B cells have a distinct phenotype, described as innate-like and weakly auto-reactive. Increased surface levels of IgM and the complement receptor CD21 may lower their activation threshold [28]. MZ B cells are efficient presenters of antigen to naïve CD4+ T cells [29]. Over-representation of this B cell subpopulation occurs independent of disease progression, and has been mapped to chromosome 4 (Idd9/11) [26]. It has been suggested the MZ enlargement in NOD is the result of defects in B cell migration within lymphoid tissues [26]. Although its genetic basis and disease relevance is unknown, CD19 expression is also modestly increased on NOD B cells [30]. CD19 is an important BCR co-receptor and accessory signal transducer that participates in Lyn tyrosine kinase and PI3-kinase activation following antigen stimulation. Heightened CD19 levels have been linked previously to B cell hyper-responsive and autoimmunity [31].
4. In NOD mice, B cells contribute to T1D development via mechanisms distinct from autoantibody production
Although a role for transplacentally acquired anti-insulin antibodies in T1D development in NOD mice has been suggested, it is unlikely that these autoantigenic antibodies make a major contribution to pathogenesis [32]. NOD mice in which B cells express membrane-bound but not secreted IgM develop T1D with penetrance comparable to wild-type NOD [33]. However, Silva et al. showed that anti-islet antibodies are capable, admittedly under very contrived circumstances, of supporting the survival and proliferation of islet-reactive CD4+ T cells. Diabetes resistant B10.BR mice were bred with a TCR transgene from clone 3A9, recognizing a dominant HEL peptide. Mice were also transgenic for the HEL transgene, with expression of soluble HEL driven by the gene promoter. Spontaneous T1D incidence, with increased thymic production of islet reactive CD4+ T cells recognizing a single experimentally expressed autoantigen, was low (<25%). Passive transfer of anti-HEL autoantibodies raised disease incidence (>70%). Increased proliferation of CFSE-labeled islet-reactive CD4+ T cells was also observed (80% CFSE diluted as opposed to 20% in the control) [34]. Despite this demonstration of principle, it seems most likely that in NOD mice B cells promote beta cell destruction and disease progression primarily through presentation of antigen to auto-reactive T cells and the secretion of pro-inflammatory cytokines.
B cells capture beta cell autoantigen via their BCR, then process and present it in peptide form to T cells. B cell-specific deletion of the MHC class II I-Ag7 prevents T1D in NOD [35]. In mixed bone marrow chimeras where only B cells are MHC class I deficient, disease development was prevented [36]. Thus, B cells likely play a role in T1D pathogenesis via interactions with both CD4+ and CD8+ T cells. Pancreas infiltrating B cells have elevated levels MHC I and II. B7-1 and B7-2 are also up regulated [37]. These B cells are primed for destructive APC function within the niche [36].
5. Antigenic targets in NOD T1D
Although antibodies reactive with multiple islet antigens can be detected in the serum of patients at high risk for T1D development [4,38,39], NOD mice have thus far been shown to only produce anti-insulin autoantibodies [40]. It is unlikely that these antibodies make a major contribution to disease [33], though their existence is indicative of a breech in B cell tolerance and ongoing antigen-specific autoimmune response. Moreover, NOD mice that express an irrelevant B cell receptor by transgenesis are resistant to T1D [9].
In NOD, insulin autoantibodies are detectable from the earliest stages of insulitis, 4 to 8 weeks of age [41]. In mice transgenic for the 125Tg heavy chain (VH125.NOD) 1–2 percent of B cells are insulin specific and transgenesis results in development of T1D at an accelerated rate. Conversely, VH281.NOD mice, whose transgenic immunoglobulin heavy chain biases the repertoire against insulin reactivity, are largely resistant to disease [15]. In our colony disease develops in <1% of VH281.NOD, and disease is only seen in mice >30 weeks of age. Disease in these mice is associated with loss of heavy chain exclusion consistent with appearance of insulin-specific B cells expressing endogenous heavy chains (unpublished observations). These B cells may thus drive disease. Indeed, in vitro, Kendall et al. showed that insulin-binding 125Tg B cells are more competent APC than their non insulin-binding counterparts [42]. Finally, the very fact that insulin-specific T cells transfer disease, implicates function of antigen-presenting insulin specific B cells in pathogenesis.
Transgenic T and B cell models specific for other pancreatic antigen targets have yet to be developed. Stadinski et al. have identified chromogranin A (ChgA) as the antigen for several highly diabetogenic T cell clones [43]. Islet amyloid polypeptide is likewise a known target antigen for disease inducing CD4+ T cells [44]. B cells specific for either of these autoantigens in the NOD have yet to be identified. Their existence, or lack thereof, would shed further light on the role of other islet antigens in disease development.
6. Efftcacy of B cell–targeted therapies in NOD
Multiple approaches have been taken to prevent or treat T1D by targeting B cells. These have included targeting of B cells for destruction using antibodies recognizing CD20 [1,2] or CD22 [45], and starvation from the critical survival factor BAFF using BCMA-Fc [46] or anti-BAFF mAb [47]. In all cases, some degree of disease prevention or reversal was achieved, but results were dependent upon timing and kinetics of B cell depletion, as well as the subsets targeted. In new onset diabetic NOD, long term disease reversal was observed in a third of mice treated with anti-CD20 [1]. However, a potential pitfall of the drug was discovered. Murine B cells down-regulate surface expression of CD20 upon entering the pancreas [48]. Thus the most pathogenic B cells may survive and continue to provide cognate help to self-reactive effector CD4+ T cells. Whether this is true of the pancreas infiltrating B cells in human T1D remains to be established.
The efficacy of B cell depleting strategies in NOD depended largely on disease state at the time of treatment. When therapy was initiated prior to the onset of insulitis disease was delayed, but not prevented with both anti-CD20 and anti-BAFF mAb [1,47]. After treatment the B cell compartment was replenished, and disease ensued. However, if treatment was begun after initiation of insulitis, in the period of elevated blood glucose (160–200 mg/dl) before overt disease (>250 mg/dl), long-lasting protection was achieved using either BCMA-Fc or anti-BAFF mAb. Disease was disrupted for the duration of each of these studies [46,47].
Pan-B cell targeted therapeutic approaches are unlikely to be accepted for general use in the clinic because of their greater risk relative to current alternatives. However, specific targeting of B cells that recognize islet antigens may be useful. To model such approaches Henry et al. attempted to affect disease development in the VH125.NOD model by treatment of animals with anti-insulin mAbs that recognize an epitope that is not sterically hindered by antigen binding to receptors containing the VH125 heavy chain [15]. The study explored was whether such antibodies would, by depleting insulin auto-reactive B cells, prevent disease development. Findings suggested this strategy could be effective despite the fact that in this study the treatment did not lead to complete depletion of the insulin-binding B cells. It is noteworthy that anecdotal evidence from many laboratories has shown that BCR-targeted antibodies are poor inducers of B cell depletion in vivo, likely because these receptors recycle rapidly, internalizing their cargo, thus preventing ADCC or complement mediated killing.
7. Efftcacy of Rituximab therapy in T1D in man
Rituximab, an anti-human CD20 monoclonal antibody that depletes most B cells in the body, was initially approved for the treatment of non-Hodgkin’s lymphoma [49]. Subsequently it been used to treat over 18 autoimmune disorders, including T1D [50]. In a TrialNet phase II clinical study, four injections of Rituximab were given to T1D patients within 3 months of diagnosis [3]. In patients given the drug, beta cell function was preserved at one year following initiation of treatment as measured by increased C-peptide levels and reduced requirement for insulin [3]. Despite the fact that significant improvement was not seen two years after treatment, this study demonstrates a role for B cells in the pathogenesis of T1D and the potential therapeutic utility of targeting of B cells. In addition, timing may be crucial for targeting of these cells, since by the time patients are hyperglycaemic, B cells may have likely already undertaken their deleterious acts as APCs in the pancreas and pancreatic lymph nodes.
8. T1D autoantigens in man
The majority of target antigens in human T1D have been identified by analysis of specificity of antibodies produced by patients. Four major autoantigens have been discovered using this approach, including: insulin, glutamic acid decarboxylase 65 (GAD65), insulinoma-associated antigen 2 (I-A2, ICA512), and the more recently described zinc transporter 8 (ZnT8) [4,51]. Although antibodies to these autoantigens are likely non-pathogenic, they have great value for identification of high-risk individuals. Anti-insulin antibodies are typically first to develop during the course of disease and occur in 90% of patients 5 years of age and younger who develop T1D [39,52,53]. In addition, half of the T cells isolated from pancreatic lymph nodes from diabetic patients recognize insulin A chain epitopes [54]. Although insulin specific B cells may play a predominant role, the tolerance/ignorance of GAD65, I-A2 and ZnT8 must also be subverted during disease development.
9. Phenotypic changes in the B cells associated with T1D in the human
The mechanism(s) by which B cells participate in development of T1D in man is relatively unstudied. However, it has been hypothesized that, as in NOD, B cells promote pathogenesis by antigen presentation. DR3/4-DQ2/8 alleles of HLA class II genes confer greatest risk of T1D development, indicating a critical role for antigen presentation to CD4 T cells [55]. HLA Class I alleles rank second among risk alleles. Interestingly, among lymphoid cells in pancreata of newly diagnosed T1D patients B cells occur in frequency second only to CD8 T cells [56]. It is tempting to speculate that these B cells function as the primary APCs for CD8 T cells, as has been shown in NOD mice [36]. These B cells must have escaped normal silencing mechanisms and responded to antigen. In this context, it has been reported that both T1D and SLE patients have decreased recombining sequence (RS) rearrangements in λ+ B cells compared to healthy controls, indicating that reduced receptor editing occurs in these autoimmune patients [57]. These findings suggest that autoimmune patients may have a higher autoreactivity threshold for initiation of RS rearrangements, which may allow more autoreactive B cells to enter the periphery. In this context it was recently shown that the Ptpn22 T1D risk allele acts in a B cell intrinsic fashion to allow autoreactive B cells to enter the periphery [58,59]. Thus circumstantial evidence supports the possibility that in human T1D islet antigen reactive B cells escape tolerance and enter the periphery, localizing in the pancreas where they promote disease by presenting antigen to CD4 and CD8 T cells.
In our own studies, we have begun to analyze the status of insulin-autoreactive B cells along the continuum of T1D development. We found that all pre-diabetic and newly diagnosed T1D patients, and some first degree relatives, exhibit reduced frequencies of both insulin-specific and total anergic BND B cells in their peripheral blood compared to long term diabetics and healthy controls (Smith and Cambier, submitted for publication). BND anergic B cells were first defined in healthy individuals as having normal levels of surface IgD but reduced levels of IgM, and were termed BND cells [60]. Greater than 75% of BND cells are autoreactive and these cells are unresponsive to BCR stimulation. These studies suggest that anergic B are transiently lost prior to development of T1D, a likely consequence of injury and/or infection. Alternatively or additionally, this loss of anergic B cells from blood may indicate that these cells have entered tissues rich in autoantigen, such as the pancreas, where they can become activated by antigen and present antigen to T cells.
10. Conclusion
Compelling evidence documents the important role for islet antigen-reactive B cells in development of type I diabetes in mouse models and in man. However, therapeutic strategies that deplete all B cells, while effective, are unlikely to find utility in the clinic due to risk considerations. Rituxan elimination of naive B cells required to mount protective immune responses to newly encountered pathogens can render patients vulnerable to infection for many months [50,61]. Targeted depletion of islet antigen, especially insulin reactive B cell may provide an effective and low risk alternative.
References
- 1.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;117:3857–67. doi: 10.1172/JCI32405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Xiu Y, Wong CP, Bouaziz JD, Hamaguchi Y, Wang Y, Pop SM, et al. B lymphocyte depletion by CD20 monoclonal antibody prevents diabetes in nonobese diabetic mice despite isotype-specific differences in Fc gamma R effector functions. J Immunol. 2008;180:2863–75. doi: 10.4049/jimmunol.180.5.2863. [DOI] [PubMed] [Google Scholar]
- 3.Pescovitz MD, Greenbaum CJ, Krause-Steinrauf H, Becker DJ, Gitelman SE, Goland R, et al. Rituximab, B-lymphocyte depletion, and preservation of beta-cell function. N Engl J Med. 2009;361:2143–52. doi: 10.1056/NEJMoa0904452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Orban T, Sosenko JM, Cuthbertson D, Krischer JP, Skyler JS, Jackson R, et al. Pancreatic islet autoantibodies as predictors of type 1 diabetes in the diabetes prevention trial-type 1. Diabetes Care. 2009;32:2269–74. doi: 10.2337/dc09-0934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Verge CF, Stenger D, Bonifacio E, Colman PG, Pilcher C, Bingley PJ, et al. Combined use of autoantibodies (IA-2 autoantibody, GAD autoantibody, insulin autoantibody, cytoplasmic islet cell antibodies) in type 1 diabetes: Combinatorial Islet Autoantibody Workshop. Diabetes. 1998;47:1857–66. doi: 10.2337/diabetes.47.12.1857. [DOI] [PubMed] [Google Scholar]
- 6.Falcone M, Lee J, Patstone G, Yeung B, Sarvetnick N. B lymphocytes are crucial antigen-presenting cells in the pathogenic autoimmune response to GAD65 antigen in nonobese diabetic mice. J Immunol. 1998;161:1163–8. [PubMed] [Google Scholar]
- 7.Greeley SA, Moore DJ, Noorchashm H, Noto LE, Rostami SY, Schlachterman A, et al. Impaired activation of islet-reactive CD4 T cells in pancreatic lymph nodes of B cell-deficient nonobese diabetic mice. J Immunol. 2001;167:4351–7. doi: 10.4049/jimmunol.167.8.4351. [DOI] [PubMed] [Google Scholar]
- 8.Serreze DV, Fleming SA, Chapman HD, Richard SD, Leiter EH, Tisch RM. B lymphocytes are critical antigen-presenting cells for the initiation of T cell-mediated autoimmune diabetes in nonobese diabetic mice. J Immunol. 1998;161:3912–8. [PubMed] [Google Scholar]
- 9.Silveira PA, Johnson E, Chapman HD, Bui T, Tisch RM, Serreze DV. The preferential ability of B lymphocytes to act as diabetogenic APC in NOD mice depends on expression of self-antigen-specific immunoglobulin receptors. Eur J Immunol. 2002;32:3657–66. doi: 10.1002/1521-4141(200212)32:12<3657::AID-IMMU3657>3.0.CO;2-E. [DOI] [PubMed] [Google Scholar]
- 10.Tian J, Zekzer D, Hanssen L, Lu Y, Olcott A, Kaufman DL. Lipopolysaccharide-activated B cells down-regulate Th1 immunity and prevent autoimmune diabetes in nonobese diabetic mice. J Immunol. 2001;167:1081–9. doi: 10.4049/jimmunol.167.2.1081. [DOI] [PubMed] [Google Scholar]
- 11.Harris DP, Haynes L, Sayles PC, Duso DK, Eaton SM, Lepak NM, et al. Reciprocal regulation of polarized cytokine production by effector B and T cells. Nat Immunol. 2000;1:475–82. doi: 10.1038/82717. [DOI] [PubMed] [Google Scholar]
- 12.Serreze DV, Chapman HD, Varnum DS, Hanson MS, Reifsnyder PC, Richard SD, et al. 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 Med. 1996;184:2049–53. doi: 10.1084/jem.184.5.2049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Vong AM, Daneshjou N, Norori PY, Sheng H, Braciak TA, Sercarz EE, et al. Spectratyping analysis of the islet-reactive T cell repertoire in diabetic NOD Igmu(null) mice after polyclonal B cell reconstitution. J Transl Med. 2011;9:101. doi: 10.1186/1479-5876-9-101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.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. Diabetes. 1997;46:941–6. doi: 10.2337/diab.46.6.941. [DOI] [PubMed] [Google Scholar]
- 15.Hulbert C, Riseili B, Rojas M, Thomas JW. B cell specificity contributes to the outcome of diabetes in nonobese diabetic mice. J Immunol. 2001;167:5535–8. doi: 10.4049/jimmunol.167.10.5535. [DOI] [PubMed] [Google Scholar]
- 16.Yang M, Charlton B, Gautam AM. Development of insulitis and diabetes in B cell-deficient NOD mice. J Autoimmun. 1997;10:257–60. doi: 10.1006/jaut.1997.0128. [DOI] [PubMed] [Google Scholar]
- 17.Peterson JD, Haskins K. Transfer of diabetes in the NOD-scid mouse by CD4 T-cell clones. Differential requirement for CD8 T-cells. Diabetes. 1996;45:328–36. doi: 10.2337/diab.45.3.328. [DOI] [PubMed] [Google Scholar]
- 18.Martin S, Wolf-Eichbaum D, Duinkerken G, Scherbaum WA, Kolb H, Noordzij JG, et al. Development of type 1 diabetes despite severe hereditary B-lymphocyte deficiency. N Engl J Med. 2001;345:1036–40. doi: 10.1056/NEJMoa010465. [DOI] [PubMed] [Google Scholar]
- 19.Broides A, Yang W, Conley ME. Genotype/phenotype correlations in X-linked agammaglobulinemia. Clin Immunol. 2006;118:195–200. doi: 10.1016/j.clim.2005.10.007. [DOI] [PubMed] [Google Scholar]
- 20.Cambier JC, Gauld SB, Merrell KT, Vilen BJ. B-cell anergy: from transgenic models to naturally occurring anergic B cells. Nat Rev Immunol. 2007;7:633–43. doi: 10.1038/nri2133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Gauld SB, Merrell KT, Cambier JC. Silencing of autoreactive B cells by anergy: a fresh perspective. Curr Opin Immunol. 2006;18:292–7. doi: 10.1016/j.coi.2006.03.015. [DOI] [PubMed] [Google Scholar]
- 22.Goodnow CC, Crosbie J, Adelstein S, Lavoie TB, Smith-Gill SJ, Brink RA, et al. Altered immunoglobulin expression and functional silencing of self-reactive B lymphocytes in transgenic mice. Nature. 1988;334:676–82. doi: 10.1038/334676a0. [DOI] [PubMed] [Google Scholar]
- 23.Gauld SB, Benschop RJ, Merrell KT, Cambier JC. Maintenance of B cell anergy requires constant antigen receptor occupancy and signaling. Nat Immunol. 2005;6:1160–7. doi: 10.1038/ni1256. [DOI] [PubMed] [Google Scholar]
- 24.Silveira PA, Dombrowsky J, Johnson E, Chapman HD, Nemazee D, Serreze DV. B cell selection defects underlie the development of diabetogenic APCs in nonobese diabetic mice. J Immunol. 2004;172:5086–94. doi: 10.4049/jimmunol.172.8.5086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Cox SL, Stolp J, Hallahan NL, Counotte J, Zhang W, Serreze DV, et al. Enhanced responsiveness to T-cell help causes loss of B-lymphocyte tolerance to a beta-cell neo-self-antigen in type 1 diabetes prone NOD mice. Eur J Immunol. 2010;40:3413–25. doi: 10.1002/eji.201040817. [DOI] [PubMed] [Google Scholar]
- 26.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. J Immunol. 2005;174:4821–7. doi: 10.4049/jimmunol.174.8.4821. [DOI] [PubMed] [Google Scholar]
- 27.Wither JE, Roy V, Brennan LA. Activated B cells express increased levels of costimulatory molecules in young autoimmune NZB and (NZB × NZW)F(1) mice. Clin Immunol. 2000;94:51–63. doi: 10.1006/clim.1999.4806. [DOI] [PubMed] [Google Scholar]
- 28.Martin F, Oliver AM, Kearney JF. Marginal zone and B1 B cells unite in the early response against T-independent blood-borne particulate antigens. Immunity. 2001;14:617–29. doi: 10.1016/s1074-7613(01)00129-7. [DOI] [PubMed] [Google Scholar]
- 29.Attanavanich K, Kearney JF. Marginal zone, but not follicular B cells, are potent activators of naive CD4 T cells. J Immunol. 2004;172:803–11. doi: 10.4049/jimmunol.172.2.803. [DOI] [PubMed] [Google Scholar]
- 30.Ziegler AI, Le Page MA, Maxwell MJ, Stolp J, Guo H, Jayasimhan A, et al. The CD19 signalling molecule is elevated in NOD mice and controls type 1 diabetes development. Diabetologia. 2013;56:2659–68. doi: 10.1007/s00125-013-3038-2. [DOI] [PubMed] [Google Scholar]
- 31.Inaoki M, Sato S, Weintraub BC, Goodnow CC, Tedder TF. CD19-regulated signaling thresholds control peripheral tolerance and autoantibody production in B lymphocytes. J Exp Med. 1997;186:1923–31. doi: 10.1084/jem.186.11.1923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.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;8:399–402. doi: 10.1038/nm0402-399. [DOI] [PubMed] [Google Scholar]
- 33.Wong FS, Wen L, Tang M, Ramanathan M, Visintin I, Daugherty J, et al. Investigation of the role of B-cells in type 1 diabetes in the NOD mouse. Diabetes. 2004;53:2581–7. doi: 10.2337/diabetes.53.10.2581. [DOI] [PubMed] [Google Scholar]
- 34.Silva DG, Daley SR, Hogan J, Lee SK, Teh CE, Hu DY, et al. Anti-islet autoantibodies trigger autoimmune diabetes in the presence of an increased frequency of islet-reactive CD4 T cells. Diabetes. 2011;60:2102–11. doi: 10.2337/db10-1344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Noorchashm H, Lieu YK, Noorchashm N, Rostami SY, Greeley SA, Schlachterman A, et al. I-Ag7-mediated antigen presentation by B lymphocytes is critical in overcoming a checkpoint in T cell tolerance to islet beta cells of nonobese diabetic mice. J Immunol. 1999;163:743–50. [PubMed] [Google Scholar]
- 36.Marino E, Tan B, Binge L, Mackay CR, Grey ST. B-cell cross-presentation of autologous antigen precipitates diabetes. Diabetes. 2012;61:2893–905. doi: 10.2337/db12-0006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Hussain S, Delovitch TL. Dysregulated B7-1 and B7-2 expression on nonobese diabetic mouse B cells is associated with increased T cell costimulation and the development of insulitis. J Immunol. 2005;174:680–7. doi: 10.4049/jimmunol.174.2.680. [DOI] [PubMed] [Google Scholar]
- 38.Gianani R, Eisenbarth GS. The stages of type 1A diabetes: 2005. Immunol Rev. 2005;204:232–49. doi: 10.1111/j.0105-2896.2005.00248.x. [DOI] [PubMed] [Google Scholar]
- 39.Vehik K, Beam CA, Mahon JL, Schatz DA, Haller MJ, Sosenko JM, et al. Development of autoantibodies in the TrialNet Natural History Study. Diabetes Care. 2011 doi: 10.2337/dc11-0560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.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: identification of insulin but not glutamic acid decarboxylase or IA-2 as specific autoantigens of humoral autoimmunity in nonobese diabetic mice. Diabetes. 2001;50:2451–8. doi: 10.2337/diabetes.50.11.2451. [DOI] [PubMed] [Google Scholar]
- 41.Yu L, Robles DT, Abiru N, Kaur P, Rewers M, Kelemen K, et al. Early expression of antiinsulin autoantibodies of humans and the NOD mouse: evidence for early determination of subsequent diabetes. Proc Natl Acad Sci USA. 2000;97:1701–6. doi: 10.1073/pnas.040556697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Kendall PL, Case JB, Sullivan AM, Holderness JS, Wells KS, Liu E, et al. Tolerant anti-insulin B cells are effective APCs. J Immunol. 2013;190:2519–26. doi: 10.4049/jimmunol.1202104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.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;11:225–31. doi: 10.1038/ni.1844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Delong T, Baker RL, Reisdorph N, Reisdorph R, Powell RL, Armstrong M, et al. Islet amyloid polypeptide is a target antigen for diabetogenic CD4+ T cells. Diabetes. 2011;60:2325–30. doi: 10.2337/db11-0288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Fiorina P, Vergani A, Dada S, Jurewicz M, Wong M, Law K, et al. Targeting CD22 reprograms B-cells and reverses autoimmune diabetes. Diabetes. 2008;57:3013–24. doi: 10.2337/db08-0420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Marino E, Villanueva J, Walters S, Liuwantara D, Mackay F, Grey ST. CD4(+)CD25(+) T-cells control autoimmunity in the absence of B-cells. Diabetes. 2009;58:1568–77. doi: 10.2337/db08-1504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Zekavat G, Rostami SY, Badkerhanian A, Parsons RF, Koeberlein B, Yu M, et al. In vivo BLyS/BAFF neutralization ameliorates islet-directed autoimmunity in nonobese diabetic mice. J Immunol. 2008;181:8133–44. doi: 10.4049/jimmunol.181.11.8133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Serreze DV, Chapman HD, Niens M, Dunn R, Kehry MR, Driver JP, et al. Loss of intra-islet CD20 expression may complicate efficacy of B-cell-directed type 1 diabetes therapies. Diabetes. 2011;60:2914–21. doi: 10.2337/db11-0705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Eisenberg R, Looney RJ. The therapeutic potential of anti-CD20 what do B-cells do? Clin Immunol. 2005;117:207–13. doi: 10.1016/j.clim.2005.08.006. [DOI] [PubMed] [Google Scholar]
- 50.Gurcan HM, Keskin DB, Stern JN, Nitzberg MA, Shekhani H, Ahmed AR. A review of the current use of rituximab in autoimmune diseases. Int Immunopharmacol. 2009;9:10–25. doi: 10.1016/j.intimp.2008.10.004. [DOI] [PubMed] [Google Scholar]
- 51.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 USA. 2007;104:17040–5. doi: 10.1073/pnas.0705894104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Williams AJ, Norcross AJ, Dix RJ, Gillespie KM, Gale EA, Bingley PJ. The prevalence of insulin autoantibodies at the onset of Type 1 diabetes is higher in males than females during adolescence. Diabetologia. 2003;46:1354–6. doi: 10.1007/s00125-003-1197-2. [DOI] [PubMed] [Google Scholar]
- 53.Vardi P, Ziegler AG, Mathews JH, Dib S, Keller RJ, Ricker AT, et al. Concentration of insulin autoantibodies at onset of type I diabetes. Inverse log-linear correlation with age. Diabetes Care. 1988;11:736–9. doi: 10.2337/diacare.11.9.736. [DOI] [PubMed] [Google Scholar]
- 54.Kent SC, Chen Y, Bregoli L, Clemmings SM, Kenyon NS, Ricordi C, et al. Expanded T cells from pancreatic lymph nodes of type 1 diabetic subjects recognize an insulin epitope. Nature. 2005;435:224–8. doi: 10.1038/nature03625. [DOI] [PubMed] [Google Scholar]
- 55.Erlich H, Valdes AM, Noble J, Carlson JA, Varney M, Concannon P, et al. HLA DR-DQ haplotypes and genotypes and type 1 diabetes risk: analysis of the type 1 diabetes genetics consortium families. Diabetes. 2008;57:1084–92. doi: 10.2337/db07-1331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Willcox A, Richardson SJ, Bone AJ, Foulis AK, Morgan NG. Analysis of islet inflammation in human type 1 diabetes. Clin Exp Immunol. 2009;155:173–81. doi: 10.1111/j.1365-2249.2008.03860.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Panigrahi AK, Goodman NG, Eisenberg RA, Rickels MR, Naji A, Luning Prak ET. RS rearrangement frequency as a marker of receptor editing in lupus and type 1 diabetes. J Exp Med. 2008;205:2985–94. doi: 10.1084/jem.20082053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Dai X, James RG, Habib T, Singh S, Jackson S, Khim S, et al. A disease-associated PTPN22 variant promotes systemic autoimmunity in murine models. J Clin Invest. 2013;123:2024–36. doi: 10.1172/JCI66963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Habib T, Funk A, Rieck M, Brahmandam A, Dai X, Panigrahi AK, et al. Altered B cell homeostasis is associated with type I diabetes and carriers of the PTPN22 allelic variant. J Immunol. 2012;188:487–96. doi: 10.4049/jimmunol.1102176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Duty JA, Szodoray P, Zheng NY, Koelsch KA, Zhang Q, Swiatkowski M, et al. Functional anergy in a subpopulation of naive B cells from healthy humans that express autoreactive immunoglobulin receptors. J Exp Med. 2009;206:139–51. doi: 10.1084/jem.20080611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Popa C, Leandro MJ, Cambridge G, Edwards JC. Repeated B lymphocyte depletion with rituximab in rheumatoid arthritis over 7 yrs. Rheumatology (Oxford) 2007;46:626–30. doi: 10.1093/rheumatology/kel393. [DOI] [PubMed] [Google Scholar]
