Skip to main content
Erschienen in: Seminars in Immunopathology 4/2008

Open Access 01.12.2008 | Review

Central tolerance: what have we learned from mice?

verfasst von: Tom M. McCaughtry, Kristin A. Hogquist

Erschienen in: Seminars in Immunopathology | Ausgabe 4/2008

Abstract

Producing a healthy immune system capable of defending against pathogens, while avoiding autoimmunity, is dependent on thymic selection. Positive selection yields functional T cells that have the potential to recognize both self and foreign antigens. Therefore, negative selection exists to manage potentially self-reactive cells. Negative selection results from the induction of anergy, receptor editing, clonal diversion (agonist selection), and/or clonal deletion (apoptosis) in self-reactive clones. Clonal deletion has been inherently difficult to study because the cells of interest are undergoing apoptosis and being eliminated quickly. Furthermore, analysis of clonal deletion in humans has proved even more difficult due to availability of samples and lack of reagents. Mouse models have thus been instrumental in achieving our current understanding of central tolerance, and the evolution of elegant model systems has led to an explosion of new data to be assimilated. This review will focus on recent advances in the field of clonal deletion with respect to three aspects: the development of physiological model systems, signaling pathways that lead to apoptosis, and antigen presenting cell types involved in the induction of clonal deletion.
The prevention of autoimmunity is primarily the outcome of central tolerance, which is achieved when thymocytes with high affinity for self-peptide/major histocompatibility complex (self-p/MHC) undergo negative selection. Four outcomes of negative selection have been described. First, receptor editing is a process by which thymocytes with high affinity for self-p/MHC are instructed to generate a second rearrangement of the T cell receptors (TCR)α loci, thereby altering the specificity of the TCR [14]. Secondly, anergy, or a state of induced unresponsiveness, has also been described [5]. The relative contribution of receptor editing and anergy to central tolerance is thought to be minimal. Instead, clonal diversion (agonist selection) of high-affinity thymocytes into lineages that attain immunoregulatory function is very important (reviewed in [69]). Finally, clonal deletion (induction of apoptosis in self-reactive clones) is the predominate mechanism by which central tolerance is achieved and will be the focus of this review.

Model systems

In 1957, Sir Macfarlane Burnet first proposed the concept of “repertoire purging” as a mechanism of lymphocyte tolerance [10]. This process was first described experimentally for thymocytes by studying clonal deletion in response to superantigens [11] (see Table 1). Superantigens are molecular remnants of proviruses in the murine genome that cross-link particular Vβ segments of TCRs with class II MHC. While this mimics a high-affinity TCR ligation, it is unclear if the downstream signaling events are entirely the same as stimulation by p/MHC. Soon after, in vitro stimulation of thymocytes with high-affinity ligands was utilized to model clonal deletion [1214]. However, it is unlikely that in vitro stimulation recapitulates the in vivo process of clonal deletion because the intact thymic microenvironment is missing. The switch to in vivo models was a substantial improvement and first utilized the injection of TCR-cross-linking antibodies to simulate high-affinity TCR ligation [15]. However, it was more recently observed that activation of peripheral T cells and their subsequent release of pro-inflammatory cytokines and stress hormones [16] could be obscuring TCR-induced death of DP thymocytes. The development of TCR transgenic mice was a major advancement in the field of thymic development and allowed researchers to examine negative selection in response to true p/MHC stimulation. Some studies have utilized exogenous administration of peptide to induce negative selection [17], but this also has the caveat of activation of peripheral T cells [18, 19]. A more physiological model has been to examine the response to endogenous high-affinity peptides, either produced transgenically (neo-self-antigens) [4, 20, 21] or naturally occurring [22, 23].
Table 1
A summary of model systems used to study negative selection
Model system
Caveats
Advantages
Disadvantages
Superantigens
Only particular Vβ TCR are deleted
Endogenous antigen
Independent of TCR affinity for p/MHC
Expression mostly in medulla
TCR signal may be qualitatively or quantitatively distinct
In vitro
Costimulation required
Technically simple
Intact thymic microenvironment lacking
αCD3/αCD28
TCR Tg peptide
In vivo: αCD3/αCD28
 
Intact in vivo environment
Activation of peripheral T cells causes nonspecific deletion. Glucocorticoid mediated
In vivo: TCR Tg with injected peptide
 
Intact in vivo environment
Activation of peripheral T cells causes nonspecific deletion. Cytokine mediated
In vivo: TCR Tg with transgenic neo-self antigen
 
Intact in vivo environment
Early TCR expression
TCR expression level higher than normal on thymic precursors
High precursor frequency/monoclonal
Transgenic-antigen artifacts (expression pattern/level)
In vivo: TCR Tg with endogenous self-antigen
 
Intact in vivo environment
Early TCR expression
TCR expression level higher than normal on thymic precursors
High precursor frequency/monoclonal
In vivo: TCR Tg mixed bone marrow chimeras with endogenous self-antigen
 
Intact in vivo environment
Early TCR expression
Precursor frequency is lower
TCR expression level higher than normal on thymic precursors
In vivo: “On-time” TCR Tg with endogenous self-antigen
 
Intact in vivo environment
TCR expression somewhat higher than normal on thymic precursors
Appropriate TCR timing (can use in mixed chimeras to obtain low precursor frequency)
In vivo: Vβ transgenics with endogenous self or neo-self antigens
 
Intact in vivo environment
Inability to track cells prior to selection in DP thymocytes
Appropriate TCR timing and level
Precursor frequency is lower/oligoclonal
The use of different model systems has led to conflicting conclusions about the developmental stage at which thymocytes undergo clonal deletion. For example, superantigen studies have suggested that deletion occurs at the single positive (SP) stage, whereas the examination of TCR transgenics and endogenous self-antigens have suggested that deletion occurs at the transition from double negative (DN) to double positive (DP). This apparent discrepancy can be partially rectified by observation that superantigens are primarily expressed in the medulla, which is the site where SP thymocytes reside. Furthermore, the nature of transgenic TCR expression has called into question the deletion observed at the DN to DP transition. Wild type thymocytes rearrange their TCRβ loci at the DN stage and if successful, they transition to the DP stage and commence rearrangement of the TCRα loci. Thus, thymocytes are not competent to undergo deletion until they have expressed a heterodimeric TCRαβ at the DP stage. However, TCR transgenic thymocytes express both TCRα and TCRβ chains early at the DN stage and probably undergo negative selection prematurely, which could explain the observation that deletion happens at the DN to DP transition. It is clear that the nature of TCR and self-antigen expression can dramatically impact the timing of clonal deletion and perhaps the molecular mechanism by which apoptosis is induced. It is therefore important to move forward by making use of the most physiological tools available.
As an example, our laboratory recently generated a TCR transgenic mouse that recapitulates the appropriate timing of TCRα expression at the DP stage (the HYcd4 model) [24]. HYcd4 thymocytes bear the same TCR as the original HY transgenic mouse made by Harold Von Boehmer’s laboratory [23] and has a high affinity for a self-peptide derived from the Y chromosome in male mice, but because of the appropriately timed expression of the HY TCRα chain at the DP stage, they undergo deletion at the DP to SP transition. This is in stark contrast to when the TCRα chain is expressed prematurely in DN thymocytes and deletion occurs at the DN to DP transition (either in the conventional HY mouse [23] or when we utilized lck-driven Cre [24]). Other studies have utilized TCRβ-only transgenic mice to eliminate nonphysiologic artifacts of transgenic TCRα expression [21, 25]. This approach has the added benefit of producing an oligoclonal repertoire with reduced precursor frequency of antigen-specific thymocytes, which also corrects defects in thymic architecture that are typical of conventional TCR transgenics [26, 27]. The latter study by Gallegos and Bevan also made use of the unique expression of tissue-restricted antigens (TRAs) in the medulla (explained in more detail below). This system obviates the artifacts introduced by premature transgenic TCR expression because only postpositive selection thymocytes, transitioning from the DP to SP stage and migrating to the medulla, are encountering high-affinity antigen and undergoing deletion (See Fig. 1). One lingering defect of all TCR transgenic models is the expression level of the TCR transgenes. Wild-type DP thymocytes express very little TCR on their cell surface and upregulate its expression following positive selection, whereas TCR transgenes driven off of strong promoters express at higher levels. Unfortunately, creating a TCRα knock-in into the endogenous locus still bears this artifact [28], although a TCRβ knock-in has not yet been created. Thus, a model for completely physiological transgenic TCR expression remains elusive. Future studies examining negative selection should strive to make use of models that have appropriately timed TCR expression, at physiological levels, with high affinity for a physiologically expressed, endogenous self-antigen. Precursor frequency of antigen-specific cells should also be minimized. Ideally, the analysis of a polyclonal repertoire should be employed; however, it is currently difficult to identify small numbers of precursors, due to the fact that DP thymocytes express low levels of TCR and are not easily distinguished by p/MHC tetramer staining.

Molecular mediators of clonal deletion

A fundamental question of T cell development has been: How does a thymocyte distinguish between positive and negative selection ligands? The pervasive “affinity model” predicts that the kinetics of TCR binding to self-p/MHC determines this outcome. There is much support for this model, and recently, the precise threshold for this distinction based on affinity was determined for class I MHC-restricted TCRs [29]. Nonetheless, how a cell translates TCR affinity into the decision between life and death is less well understood. It has been suggested that high-affinity TCR ligation induces a conformational change in the cytoplasmic tail of CD3ɛ, which exposes a polyproline sequence that serves as a binding site for the adapter protein Nck [30]. However, another study suggested that low-affinity interactions also expose the polyproline sequence, albeit less efficiently [31]. Recently, Mingueneau and colleagues created knock-in mice with a mutation in the CD3 polyproline region. Surprisingly, they found that binding of Nck does not depend on previous TCR ligation. Additionally, positive selection was affected by introduction of the mutation in two TCR transgenic models, but negative selection was not [32]. The misshapen-Nck-interacting kinase-related kinase (MINK) was also suggested to be important for the induction of negative selection [33], although this finding has not further corroborated. With respect to TCR-proximal signaling events, it has been suggested that differential activation of the extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) pathways is important in this discrimination (discussed in depth in [34]). A recent report extended these findings by demonstrating the differential subcellular localization of Ras and mitogen-activated protein kinase signaling intermediates in response to positively and negatively selecting ligands [35]. Thus, it appears that Ras-mediated activation of the ERK pathway is dispensable for clonal deletion. Interestingly, Ras signaling is not only involved in positive selection of conventional T cells [36] but also in the generation of Foxp3+ Tregs [37], which seem to be selected by high affinity ligands [6]. This may be important for the decision between clonal deletion and clonal diversion. It was also recently shown that deficiency of the protein phosphatase G5PR resulted in hyperactivation of JNK and caspase 3 [38], indicating that G5PR inhibits apoptosis in the steady state and that overcoming this inhibition by G5PR is perhaps necessary for clonal deletion. A role for costimulation to induce clonal deletion has also been suggested. However, this has been controversial, likely due to use of different model systems (see [34] for a thorough review). While αCD28 strongly costimulates apoptosis in vitro, no single costimulatory molecule has been shown to be required in all models of clonal deletion. It is possible that multiple distinct costimulatory molecules function redundantly in clonal deletion.
The signals leading to clonal deletion ultimately activate the apoptosis pathway (see [39] for a detailed review), and a number of groups have implicated the orphan nuclear steroid receptor Nur77 as being necessary [40, 41]. It was shown using cultured thymocytes that myocyte enhancer-binding factor 2D (MEF2D) and ERK5 mediate transcription of Nur77 [42]. On the other hand, Nur77 expression is repressed by histone deacetylase 7 (HDAC7), and this inhibition can be overcome by TCR stimulation that activates protein kinase D1 which subsequently phosphorylates HDAC7 and promotes its nuclear export [4345]. While it has been well documented that Nur77 is induced following a strong TCR stimulation, it is less clear how Nur77 exerts its pro-apoptotic function. It was first proposed that Nur77 acts a transcription factor to promote apoptosis [46], but a more recent report suggested that Nur77 mediates apoptosis by translocating to the mitochondria where it sequesters the anti-apoptotic molecule Bcl-2 [47]. It remains unclear if the role of Nur77 as a transcription factor is also necessary. Defining the precise role for Nur77 in negative selection has been further complicated by the observation that Nor-1, a closely related family member, can have functional redundancy with Nur77 [48].
The pro-apoptotic molecule Bim also plays a central role in clonal deletion [49]. The precise mechanisms leading to Bim induction have not been fully elucidated, although Ca2+ and protein kinase C were suggested to be important [50]. A number of recent reports have investigated the transcriptional profile of thymocytes undergoing negative selection [5155]. The most recent of these, by Baldwin and Hogquist, compared the gene array data sets generated by the different groups and found surprisingly little overlap between them. Specifically, only Bim, Nur77, PD-1, and Gadd45β were consistently found to be upregulated. This might suggest that clonal deletion only requires a small number of genes to be newly synthesized. However, it has yet to be demonstrated that new transcription of any of these genes after the initial TCR ligation is absolutely necessary for deletion. It has been suggested that transcription of Bim is required [50], but it has also been argued that phosphorylation of Bim is the critical event in promoting clonal deletion [56].
Finally, an intriguing report recently suggested that posttranscriptional regulation of mRNA by microRNAs might be important in the distinction between positive and negative selection [57]. Li et al. showed that miR1–181a acts as an intrinsic “rheostat” to control T cell sensitivity in thymocytes and mature T cells. It is likely that miR-181a plays a role in setting the bandwidth for discrimination of positive and negative selection signals, and perhaps the regulation of miR-181a itself or its targets plays a role in inducing apoptosis following a high-affinity TCR signal. Our understanding of the role of microRNA in T cell development is in its infancy and is likely to be even more greatly appreciated in the future as evidenced by other recent work examining DICER and microRNA in T cell development and function [5860].

Cell types that mediate clonal deletion

Cortical thymic epithelial cells

Thymocytes interact with a variety of antigen-presenting cells during the course of their development. DN thymocytes enter the thymus at the corticomedullary junction (CMJ) and traverse outwards towards the subcapsular region where they undergo rearrangement of the TCRβ chain (Fig. 1). As DPs, they migrate randomly though a dense matrix of cortical thymic epithelial cells (cTECs), and their engagement with p/MHC on cTECs is crucial for positive selection [61]. Whether or not cTECs are also capable of inducing negative selection is less clearly understood. A number of studies have concluded that cTECs are not capable of inducing tolerance to self-antigens [6266]. However, there is also much data to suggest that cTECs can in fact be tolerogenic [4, 6775]. This apparent contradiction is likely explained by the observation made by Goldman et al. that studies indicating the thymic epithelium was not capable of inducing tolerance “were done using antigens not normally expressed by thymic epithelium and/or targets derived from other tissues”. They then go on to suggest that cTECs are capable of inducing tolerance to antigens expressed by cTECs, but not to all antigens expressed in the body. In addition, we have shown that cTECs are inefficient at inducing apoptosis of self-reactive thymocytes but that they are ultimately tolerogenic because they prevented the development of mature SP thymocytes [76]. We were unable to determine if cTECs induced anergy or clonal diversion of these cells, but these observations may also help reconcile the apparent contradictions in previous studies because the induction of tolerance can occur by mechanisms other than clonal deletion.
An intriguing study was recently reported that potentially changes the paradigm of cTECs in positive and negative selection [77]. In addition to the “standard” proteasome and the “immunoproteasome”, Murata et al. described a novel proteasomal subunit expressed exclusively in cTECs, which they termed β5t. This “thymoproteasome” has unique peptidase activity with reduced chymotrypsin-like activity, which is important for generating peptides with hydrophobic C-termini. Hydrophobic C-termini anchor peptides in the groove of class I MHC. Therefore, it is likely that cTECs not only generate a unique peptide repertoire but also present unstable class I MHC on their cell surface. These observations may explain why cTECs are critical for positive selection, and in fact, β5t-deficient mice had a dramatic defect in generation of CD8 SPs. This unique feature of cTECs also potentially explains why cTECs are incapable of inducing tolerance to antigens expressed throughout the body and their poor ability to induce clonal deletion. It is interesting to speculate that a similar mechanism exists in cTECs for the generation of class II p/mHC complexes, perhaps via cathepsins [78, 79].

Trafficking to the medulla

The notion that cTECs are not sufficient for induction of clonal deletion is bolstered by the observation that trafficking to the medulla is necessary for achieving complete tolerance [80, 81] and that this migration is primarily mediated by CCR7-dependant signaling [82, 83] (See Fig. 1). Indeed, mice with medullar defects also display autoimmune phenotypes (reviewed in [84]). It is commonly believed that the medulla is a specialized anatomical location for clonal deletion due to the high density of dendritic cells (DC) and the peculiar ability of medullary thymic epithelial cells (mTECs) to ectopically express TRAs (both of which are described in further detail below). However, a defect in clonal deletion may not be sufficient to explain the lack of tolerance in previously mentioned studies. This is because the medulla has also been implicated to be important for the generation of immunoregulatory T cells [8588] and for conventional T cells to undergo final maturation and adjustment of their TCR sensitivity to self-antigens (a process referred to as “TCR tuning”) [89]. In addition, Reinhold Förster’s laboratory also questioned whether or not CCR7 was the only chemokine receptor involved in corticomedullar migration [90]. While there is clearly medullar dysplasia in CCR7-deficient mice and reduced medullar volume, they showed that proportionally normal numbers of CD4 SP thymocytes were found in the small pockets of medulla that remained. They concluded that a redundant mechanism exists for migration to the medulla. Importantly though, it should be noted that the majority of SPs in the medulla were CD4 SPs, and it was impossible to distinguish if these were truly developing thymocytes or another lineage of CD4+ T cells, such as Foxp3+ Tregs that also reside in the medulla [91] and may have an alternate homing mechanism.

Medullary thymic epithelial cells

Despite the complicating factors of Treg development, TCR tuning, and the role of CCR7-mediated chemotaxis, it is clear that developing thymocytes must gain access to the medulla to be screened against a panel of self-antigens uniquely expressed there (See Fig. 1). It has been shown that medullary thymic epithelial cells are capable of expressing the autoimmune regulator (AIRE), which leads to the ectopic expression of tissue-restricted antigens [92, 93]. This phenomenon of mTECs is crucial for the establishment of central tolerance as AIRE-deficient humans and mice both develop autoimmunity [94]. It has been shown that mTECs express the costimulatory molecules B7-1 and B7-2 and that direct presentation of TRAs by mTECs is sufficient to induce clonal deletion of antigen-specific thymocytes, although cross-presentation by dendritic cells also occurs [21] (discussed in further detail below).
The development of mTECs is dependent upon a population of CD4+ CD3 lymphoid tissue inducer cells and receptor activator of nuclear factor κB (RANK)–RANK ligand interactions, and their maintenance is, in part, controlled by CD40–CD40L signaling and lymphotoxin-β receptor (LTβR; see Fig. 1) [9597]. Furthermore, tumor necrosis factor receptor-associated factor 6 (TRAF6), which signals downstream of RANK and CD40, is also needed for mTEC development [86]. TRAF6 activates the transcription factor NF-κB, which is also required for mTEC development [98], as is the NF-κB-inducing kinase [85] and IκB kinase α [99].
Recent work has also demonstrated that mTECs are a highly dynamic population with high proliferative capacity and a turnover rate of about 2 weeks [100, 101]. It was observed that the majority of proliferating mTECs were mature, UEA-1+ MHC class IIhi, and that these mature mTECs are the ones expressing AIRE [102]. In addition, it appears that AIRE-expressing mTECs are short-lived with rapid turnover [103]. While AIRE-dependent TRAs are found clustered throughout the genome, implying a role for epigenetics in AIRE function [104], it has also been shown that individual mTECs express a stochastic battery of TRAs, such that each mTEC is unique with respect to its antigen-presentation profile [105, 106]. Taken together, these data paint a picture where mTECs are critical for the elimination of self-reactive thymocytes and that individual AIRE-expressing mTECs are highly diverse and turning over rapidly. As loss of tolerance to just one tissue-restricted antigen is sufficient to cause autoimmunity [107], it becomes clear that thymocytes must have ample opportunity to peruse the entire milieu of the medulla to be adequately screened for self-reactivity. We recently showed that the length of time that an SP thymocyte spends in the medulla is roughly 4–5 days [108], during which a thymocyte “tunes” its TCR and becomes refractory to apoptosis. Therefore, a short medullary residency time of SP thymocytes, in combination with the diversity and turnover of mTECs expressing TRAs, may be a weak point in the mechanism of central tolerance. Evolution has obviously created a system that prevents overt autoimmunity but bear in mind that T cell-driven autoimmunity is disturbingly common in the human population. Finally, the precise mechanism by which AIRE operates is not entirely understood. AIRE most likely operates as a transcription factor, but as mentioned above with regards to epigenetics, it remains possible that AIRE promotes ectopic expression of TRAs by another mechanism(s) [109]. In addition, not all TRAs are AIRE-dependent [104]; thus, another mechanism or molecule, similar to AIRE, must also exist, perhaps involving LTβR [110].

Dendritic cells

Dogma states that TCR activation and clonal deletion are most efficiently induced by bone marrow-derived cells, and it is widely believed that dendritic cells are the principle mediator of clonal deletion. This is because DC are thought of as “professional” antigen presenting cells with high levels of class II MHC on the cell surface as well as high levels of costimulatory molecules, such as B7-1 and B7-2. Indeed, a direct role for DC in clonal deletion has been demonstrated [21, 111, 112]. However, the heterogeneity in thymic DC is largely underappreciated and not completely understood. It is unclear if all DC in the thymus are equal in their capacity to induce clonal deletion. Work by Donskoy and Goldschneider has shown that at least two distinct populations of thymic DCs exist: one that develops intrathymically and one that is derived from peripheral DCs migrating to the thymus [113]. It was suggested that these two populations might have functional differences in their ability to induce deletion of self-reactive thymocytes vs. clonal diversion of immunoregulatory cells. Goldschneider and Cone propose a model where intrathymically derived DC mediate clonal deletion, whereas extrathymically derived cells support the agonist selection of regulatory T cells (see [114] for a comprehensive review of this argument). It is important to note that the phenotype is unknown of the subset of peripheral DCs that are capable of migrating to the thymus and mediating this selection. In line with this hypothesis, it has been shown in the human thymus that a group of mTECs called Hassall’s corpuscles produces TSLP (thymic stromal lymphopoietin) and this allows DC to induce the proliferation and differentiation of Foxp3+ Tregs [115]. Conversely, it was more recently demonstrated that antigen-loaded, splenic DC were capable of homing to the thymus and inducing deletion of antigen-specific thymocytes [116]. More detailed analysis of the phenotype of the DC migrating to the thymus was also performed, and these DC included all subsets of splenic DC that were present at the time of injection, although LPS-matured DC showed a reduced capacity to home to the thymus. It may be that the ability of peripheral DC to migrate to the thymus and induce deletion vs. agonist selection might not be mutually exclusive and may depend upon the specific environmental and experimental conditions. It is also possible that different subsets of peripheral DC are capable of mediating the different selection outcomes.
As potential support for the latter hypothesis, Ken Shortman’s laboratory has shown that, in addition to intrathymically- and extrathymically derived DCs, at least three other DC subsets can be identified based upon cell surface markers (reviewed in [117]). Their data showed an initial division of DC into two populations, 35% of which are plasmacytoid DC (pDC) and the remaining 65% are conventional DC (cDC). The distinction between these two populations is based on expression of CD11c and CD45RA, with pDC being CD11cint and CD45RA+, and cDC being CD11chi and CD45RA. The majority of pDC are Ly6c+, although a small fraction of Ly6c can also be found. Similar to pDC in the spleen and lymph node, some thymic pDC can express CD4 and/or CD8α, but unlike peripheral pDC, they express high levels of TLR7 and 9, but only low levels of TLR 2, 3, and 4. Also similar to peripheral pDC, thymic pDC express low levels of MHC class II and costimulatory molecules and can take up antigen by endocytosis but not phagocytosis. Thus, pDC are weak stimulators of T cells. On the other hand, all conventional DC are MHC class II+ and the majority are CD8αα+. Similar to the minority population of CD8+ DC in the spleen and lymph node, this population in the thymus is DEC-205+ (CD205) and CD11b. However, unlike peripheral CD8α+ DC, some thymic CD8α+ cDC also express BP-1, although no functional difference has yet been shown based upon this distinction. Conventional DC can be further subdivided by their expression of CD8α and Sirpα (CD172a). CD8αlo Sirpα+ DCs account for 20% of the thymic cDC population, and similar to splenic CD8α DC, this population is also CD11bint. While thymic cDC express the costimulatory molecules B7-1 and B7-2 at slightly higher levels than their peripheral counterparts, for the most part, they appear to be immature because they are capable of phagocytic or endocytic uptake of antigens and process and present them on MHC class II. In addition, they upregulate expression of MHC class II and costimulatory molecules following activation. It is not entirely clear how this classification of thymic DC aligns with the division created by the Goldschneider laboratory as to the origin of these cell types. It seems that most of CD8α+ cDC develop intrathymically from a lymphoid prothymocyte precursor, whereas extrathymically derived DC include the CD8α DC population, likely of myeloid origin.
Finally, it is well documented that the majority of thymic DCs are found in the medulla, and therefore, it is thought that the anatomical location of clonal deletion is at the CMJ or in the medulla. However, we and others have noted the sparse but distinct presence of DC in the cortex of the thymus (See Fig. 1). This often-overlooked observation raises the possibility that cortical DC may be sufficient to mediate clonal deletion. Indeed, we have recently shown that clonal deletion to ubiquitous self-antigens can occur in the cortex, with no involvement of the medulla [76]. Furthermore, we found that thymocytes undergoing this process were preferentially in contact with DC present in the cortex and that conditional ablation of DC significantly impaired clonal deletion of antigen-specific cells. The fact that clonal deletion to ubiquitous self-antigens can occur in the cortex does not preclude the requirement of migration to the medulla and clonal deletion to TRAs. It does, however, expand our thinking as to the mechanism underlying clonal deletion. It is unknown if any phenotypic differences exist between cortical and medullar DC, or if any subset(s) of DC discussed previously reside preferentially in the cortex or the medulla. It was recently shown that most cortical DC are found in close association with small blood capillaries that express CCR7 ligands ([118] and our unpublished data). As positively selected thymocytes also express CCR7, it is interesting to speculate that this exists as an initial mechanism for screening thymocytes for clonal deletion. It also raises the possibility that thymocyte migration to the medulla does not occur randomly but that CCR7+ cells travel along blood capillaries, which ultimately lead to the CMJ. Finally, given that cTEC are not efficient at inducing clonal deletion and DC are, it is likely that a second signal that can be delivered by a DC but not a cTEC is necessary to induce apoptosis. We have observed that cortical DCs express B7-2, and it is tempting to think that this is the second signal. However, as discussed previously, there is no absolute role for costimulation by B7-2, although it has been shown that complete deletion of superantigen-reactive cells requires B7-1/2 and CD28 interaction [119]. Therefore, it remains possible that B7-1 and B7-2 play a role in clonal deletion, but it is probable that other costimulatory molecules also exist.
In conclusion, recent advancements have allowed researchers to gain a deeper understanding of the process of clonal deletion. We are finally beginning to understand the molecular pathways that are necessary for the elimination of self-reactive thymocytes, as well as the cellular players that contribute to this process. As the field moves ahead, it is important to develop and use the most physiological tools possible and to consider the intricate anatomical microenvironments of the thymus in order to gain a clear understanding of clonal deletion.

Open Access

This article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.
Open AccessThis is an open access article distributed under the terms of the Creative Commons Attribution Noncommercial License (https://​creativecommons.​org/​licenses/​by-nc/​2.​0), which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
1.
2.
Zurück zum Zitat Santori FR, Arsov I, Lilic M, Vukmanovic S (2002) Editing autoreactive TCR enables efficient positive selection. J Immunol 169:1729–1734PubMed Santori FR, Arsov I, Lilic M, Vukmanovic S (2002) Editing autoreactive TCR enables efficient positive selection. J Immunol 169:1729–1734PubMed
3.
Zurück zum Zitat Wang F, Huang CY, Kanagawa O (1998) Rapid deletion of rearranged T cell antigen receptor (TCR) Valpha–Jalpha segment by secondary rearrangement in the thymus: role of continuous rearrangement of TCR alpha chain gene and positive selection in the T cell repertoire formation. Proc Natl Acad Sci U S A 95:11834–11839. doi:10.1073/pnas.95.20.11834 PubMed Wang F, Huang CY, Kanagawa O (1998) Rapid deletion of rearranged T cell antigen receptor (TCR) Valpha–Jalpha segment by secondary rearrangement in the thymus: role of continuous rearrangement of TCR alpha chain gene and positive selection in the T cell repertoire formation. Proc Natl Acad Sci U S A 95:11834–11839. doi:10.​1073/​pnas.​95.​20.​11834 PubMed
4.
Zurück zum Zitat Mayerova D, Hogquist KA (2004) Central tolerance to self-antigen expressed by cortical epithelial cells. J Immunol 172:851–856PubMed Mayerova D, Hogquist KA (2004) Central tolerance to self-antigen expressed by cortical epithelial cells. J Immunol 172:851–856PubMed
5.
Zurück zum Zitat Hammerling GJ, Schonrich G, Momburg F, Auphan N, Malissen M, Malissen B, Schmitt-Verhulst AM, Arnold B (1991) Non-deletional mechanisms of peripheral and central tolerance: studies with transgenic mice with tissue-specific expression of a foreign MHC class I antigen. Immunol Rev 122:47–67. doi:10.1111/j.1600-065X.1991.tb00596.x PubMed Hammerling GJ, Schonrich G, Momburg F, Auphan N, Malissen M, Malissen B, Schmitt-Verhulst AM, Arnold B (1991) Non-deletional mechanisms of peripheral and central tolerance: studies with transgenic mice with tissue-specific expression of a foreign MHC class I antigen. Immunol Rev 122:47–67. doi:10.​1111/​j.​1600-065X.​1991.​tb00596.​x PubMed
8.
Zurück zum Zitat Baldwin TA, Hogquist KA, Jameson SC (2004) The fourth way? Harnessing aggressive tendencies in the thymus. J Immunol 173:6515–6520PubMed Baldwin TA, Hogquist KA, Jameson SC (2004) The fourth way? Harnessing aggressive tendencies in the thymus. J Immunol 173:6515–6520PubMed
10.
Zurück zum Zitat Burnet FM (1957) A modification of Jerne’s theory of antibody production using the concept of clonal selection. Aust J Sci 20:67 Burnet FM (1957) A modification of Jerne’s theory of antibody production using the concept of clonal selection. Aust J Sci 20:67
12.
Zurück zum Zitat Smith CA, Williams GT, Kingston R, Jenkinson EJ, Owen JJ (1989) Antibodies to CD3/T-cell receptor complex induce death by apoptosis in immature T cells in thymic cultures. Nature 337:181–184. doi:10.1038/337181a0 PubMed Smith CA, Williams GT, Kingston R, Jenkinson EJ, Owen JJ (1989) Antibodies to CD3/T-cell receptor complex induce death by apoptosis in immature T cells in thymic cultures. Nature 337:181–184. doi:10.​1038/​337181a0 PubMed
13.
Zurück zum Zitat Jenkinson EJ, Kingston R, Smith CA, Williams GT, Owen JJ (1989) Antigen-induced apoptosis in developing T cells: a mechanism for negative selection of the T cell receptor repertoire. Eur J Immunol 19:2175–2177. doi:10.1002/eji.1830191132 PubMed Jenkinson EJ, Kingston R, Smith CA, Williams GT, Owen JJ (1989) Antigen-induced apoptosis in developing T cells: a mechanism for negative selection of the T cell receptor repertoire. Eur J Immunol 19:2175–2177. doi:10.​1002/​eji.​1830191132 PubMed
14.
Zurück zum Zitat Swat W, Ignatowicz L, von Boehmer H, Kisielow P (1991) Clonal deletion of immature CD4+8+ thymocytes in suspension culture by extrathymic antigen-presenting cells. Nature 351:150–153. doi:10.1038/351150a0 PubMed Swat W, Ignatowicz L, von Boehmer H, Kisielow P (1991) Clonal deletion of immature CD4+8+ thymocytes in suspension culture by extrathymic antigen-presenting cells. Nature 351:150–153. doi:10.​1038/​351150a0 PubMed
15.
Zurück zum Zitat Shi YF, Bissonnette RP, Parfrey N, Szalay M, Kubo RT, Green DR (1991) In vivo administration of monoclonal antibodies to the CD3 T cell receptor complex induces cell death (apoptosis) in immature thymocytes. J Immunol 146:3340–3346PubMed Shi YF, Bissonnette RP, Parfrey N, Szalay M, Kubo RT, Green DR (1991) In vivo administration of monoclonal antibodies to the CD3 T cell receptor complex induces cell death (apoptosis) in immature thymocytes. J Immunol 146:3340–3346PubMed
16.
Zurück zum Zitat Brewer JA, Kanagawa O, Sleckman BP, Muglia LJ (2002) Thymocyte apoptosis induced by T cell activation is mediated by glucocorticoids in vivo. J Immunol 169:1837–1843PubMed Brewer JA, Kanagawa O, Sleckman BP, Muglia LJ (2002) Thymocyte apoptosis induced by T cell activation is mediated by glucocorticoids in vivo. J Immunol 169:1837–1843PubMed
17.
Zurück zum Zitat Murphy KM, Heimberger AB, Loh DY (1990) Induction by antigen of intrathymic apoptosis of CD4+CD8+TCRlo thymocytes in vivo. Science (New York, NY) 250:1720–1723 Murphy KM, Heimberger AB, Loh DY (1990) Induction by antigen of intrathymic apoptosis of CD4+CD8+TCRlo thymocytes in vivo. Science (New York, NY) 250:1720–1723
18.
19.
Zurück zum Zitat Zhan Y, Purton JF, Godfrey DI, Cole TJ, Heath WR, Lew AM (2003) Without peripheral interference, thymic deletion is mediated in a cohort of double-positive cells without classical activation. Proc Natl Acad Sci U S A 100:1197–1202. doi:10.1073/pnas.0237316100 PubMed Zhan Y, Purton JF, Godfrey DI, Cole TJ, Heath WR, Lew AM (2003) Without peripheral interference, thymic deletion is mediated in a cohort of double-positive cells without classical activation. Proc Natl Acad Sci U S A 100:1197–1202. doi:10.​1073/​pnas.​0237316100 PubMed
20.
Zurück zum Zitat Baldwin KK, Trenchak BP, Altman JD, Davis MM (1999) Negative selection of T cells occurs throughout thymic development. J Immunol 163:689–698PubMed Baldwin KK, Trenchak BP, Altman JD, Davis MM (1999) Negative selection of T cells occurs throughout thymic development. J Immunol 163:689–698PubMed
21.
Zurück zum Zitat Gallegos AM, Bevan MJ (2004) Central tolerance to tissue-specific antigens mediated by direct and indirect antigen presentation. J Exp Med 200:1039–1049. doi:10.1084/jem.20041457 PubMed Gallegos AM, Bevan MJ (2004) Central tolerance to tissue-specific antigens mediated by direct and indirect antigen presentation. J Exp Med 200:1039–1049. doi:10.​1084/​jem.​20041457 PubMed
22.
Zurück zum Zitat Sha WC, Nelson CA, Newberry RD, Kranz DM, Russell JH, Loh DY (1988) Positive and negative selection of an antigen receptor on T cells in transgenic mice. Nature 336:73–76. doi:10.1038/336073a0 PubMed Sha WC, Nelson CA, Newberry RD, Kranz DM, Russell JH, Loh DY (1988) Positive and negative selection of an antigen receptor on T cells in transgenic mice. Nature 336:73–76. doi:10.​1038/​336073a0 PubMed
23.
Zurück zum Zitat Kisielow P, Bluthmann H, Staerz UD, Steinmetz M, von Boehmer H (1988) Tolerance in T-cell-receptor transgenic mice involves deletion of nonmature CD4+8+ thymocytes. Nature 333:742–746. doi:10.1038/333742a0 PubMed Kisielow P, Bluthmann H, Staerz UD, Steinmetz M, von Boehmer H (1988) Tolerance in T-cell-receptor transgenic mice involves deletion of nonmature CD4+8+ thymocytes. Nature 333:742–746. doi:10.​1038/​333742a0 PubMed
24.
Zurück zum Zitat Baldwin TA, Sandau MM, Jameson SC, Hogquist KA (2005) The timing of TCR alpha expression critically influences T cell development and selection. J Exp Med 202:111–121. doi:10.1084/jem.20050359 PubMed Baldwin TA, Sandau MM, Jameson SC, Hogquist KA (2005) The timing of TCR alpha expression critically influences T cell development and selection. J Exp Med 202:111–121. doi:10.​1084/​jem.​20050359 PubMed
25.
Zurück zum Zitat Bouneaud C, Kourilsky P, Bousso P (2000) Impact of negative selection on the T cell repertoire reactive to a self-peptide: a large fraction of T cell clones escapes clonal deletion. Immunity 13:829–840. doi:10.1016/S1074-7613(00)00080-7 PubMed Bouneaud C, Kourilsky P, Bousso P (2000) Impact of negative selection on the T cell repertoire reactive to a self-peptide: a large fraction of T cell clones escapes clonal deletion. Immunity 13:829–840. doi:10.​1016/​S1074-7613(00)00080-7 PubMed
26.
Zurück zum Zitat Canelles M, Park ML, Schwartz OM, Fowlkes BJ (2003) The influence of the thymic environment on the CD4-versus-CD8 T lineage decision. Nat Immunol 4:756–764. doi:10.1038/ni953 PubMed Canelles M, Park ML, Schwartz OM, Fowlkes BJ (2003) The influence of the thymic environment on the CD4-versus-CD8 T lineage decision. Nat Immunol 4:756–764. doi:10.​1038/​ni953 PubMed
29.
Zurück zum Zitat Naeher D, Daniels MA, Hausmann B, Guillaume P, Luescher I, Palmer E (2007) A constant affinity threshold for T cell tolerance. J Exp Med 204:2553–2559. doi:10.1084/jem.20070254 PubMed Naeher D, Daniels MA, Hausmann B, Guillaume P, Luescher I, Palmer E (2007) A constant affinity threshold for T cell tolerance. J Exp Med 204:2553–2559. doi:10.​1084/​jem.​20070254 PubMed
30.
Zurück zum Zitat Risueno RM, van Santen HM, Alarcon B (2006) A conformational change senses the strength of T cell receptor-ligand interaction during thymic selection. Proc Natl Acad Sci U S A 103:9625–9630. doi:10.1073/pnas.0601785103 PubMed Risueno RM, van Santen HM, Alarcon B (2006) A conformational change senses the strength of T cell receptor-ligand interaction during thymic selection. Proc Natl Acad Sci U S A 103:9625–9630. doi:10.​1073/​pnas.​0601785103 PubMed
31.
Zurück zum Zitat Gil D, Schrum AG, Alarcon B, Palmer E (2005) T cell receptor engagement by peptide-MHC ligands induces a conformational change in the CD3 complex of thymocytes. J Exp Med 201:517–522. doi:10.1084/jem.20042036 PubMed Gil D, Schrum AG, Alarcon B, Palmer E (2005) T cell receptor engagement by peptide-MHC ligands induces a conformational change in the CD3 complex of thymocytes. J Exp Med 201:517–522. doi:10.​1084/​jem.​20042036 PubMed
32.
Zurück zum Zitat Mingueneau M, Sansoni A, Gregoire C, Roncagalli R, Aguado E, Weiss A, Malissen M, Malissen B (2008) The proline-rich sequence of CD3epsilon controls T cell antigen receptor expression on and signaling potency in preselection CD4+CD8+ thymocytes. Nat Immunol 9:522–532. doi:10.1038/ni.1608 PubMed Mingueneau M, Sansoni A, Gregoire C, Roncagalli R, Aguado E, Weiss A, Malissen M, Malissen B (2008) The proline-rich sequence of CD3epsilon controls T cell antigen receptor expression on and signaling potency in preselection CD4+CD8+ thymocytes. Nat Immunol 9:522–532. doi:10.​1038/​ni.​1608 PubMed
33.
Zurück zum Zitat McCarty N, Paust S, Ikizawa K, Dan I, Li X, Cantor H (2005) Signaling by the kinase MINK is essential in the negative selection of autoreactive thymocytes. Nat Immunol 6:65–72. doi:10.1038/ni1145 PubMed McCarty N, Paust S, Ikizawa K, Dan I, Li X, Cantor H (2005) Signaling by the kinase MINK is essential in the negative selection of autoreactive thymocytes. Nat Immunol 6:65–72. doi:10.​1038/​ni1145 PubMed
34.
Zurück zum Zitat Palmer E (2003) Negative selection—clearing out the bad apples from the T-cell repertoire. Natl Rev 3:383–391 Palmer E (2003) Negative selection—clearing out the bad apples from the T-cell repertoire. Natl Rev 3:383–391
35.
Zurück zum Zitat Daniels MA, Teixeiro E, Gill J, Hausmann B, Roubaty D, Holmberg K, Werlen G, Hollander GA, Gascoigne NR, Palmer E (2006) Thymic selection threshold defined by compartmentalization of Ras/MAPK signalling. Nature 444:724–729. doi:10.1038/nature05269 PubMed Daniels MA, Teixeiro E, Gill J, Hausmann B, Roubaty D, Holmberg K, Werlen G, Hollander GA, Gascoigne NR, Palmer E (2006) Thymic selection threshold defined by compartmentalization of Ras/MAPK signalling. Nature 444:724–729. doi:10.​1038/​nature05269 PubMed
36.
Zurück zum Zitat Priatel JJ, Chen X, Dhanji S, Abraham N, Teh HS (2006) RasGRP1 transmits prodifferentiation TCR signaling that is crucial for CD4 T cell development. J Immunol 177:1470–1480PubMed Priatel JJ, Chen X, Dhanji S, Abraham N, Teh HS (2006) RasGRP1 transmits prodifferentiation TCR signaling that is crucial for CD4 T cell development. J Immunol 177:1470–1480PubMed
37.
Zurück zum Zitat Chen X, Priatel JJ, Chow MT, Teh HS (2008) Preferential development of CD4 and CD8 T regulatory cells in RasGRP1-deficient mice. J Immunol 180:5973–5982PubMed Chen X, Priatel JJ, Chow MT, Teh HS (2008) Preferential development of CD4 and CD8 T regulatory cells in RasGRP1-deficient mice. J Immunol 180:5973–5982PubMed
38.
Zurück zum Zitat Xing Y, Wang X, Igarashi H, Kawamoto H, Sakaguchi N (2008) Protein phosphatase subunit G5PR that regulates the JNK-mediated apoptosis signal is essential for the survival of CD4 and CD8 double-positive thymocytes. Mol Immunol 45:2028–2037. doi:10.1016/j.molimm.2007.10.028 PubMed Xing Y, Wang X, Igarashi H, Kawamoto H, Sakaguchi N (2008) Protein phosphatase subunit G5PR that regulates the JNK-mediated apoptosis signal is essential for the survival of CD4 and CD8 double-positive thymocytes. Mol Immunol 45:2028–2037. doi:10.​1016/​j.​molimm.​2007.​10.​028 PubMed
39.
Zurück zum Zitat Strasser A, Puthalakath H, O’Reilly LA, Bouillet P (2008) What do we know about the mechanisms of elimination of autoreactive T and B cells and what challenges remain. Immunol Cell Biol 86:57–66. doi:10.1038/sj.icb.7100141 PubMed Strasser A, Puthalakath H, O’Reilly LA, Bouillet P (2008) What do we know about the mechanisms of elimination of autoreactive T and B cells and what challenges remain. Immunol Cell Biol 86:57–66. doi:10.​1038/​sj.​icb.​7100141 PubMed
40.
Zurück zum Zitat Cho HJ, Edmondson SG, Miller AD, Sellars M, Alexander ST, Somersan S, Punt JA (2003) Cutting edge: identification of the targets of clonal deletion in an unmanipulated thymus. J Immunol 170:10–13PubMed Cho HJ, Edmondson SG, Miller AD, Sellars M, Alexander ST, Somersan S, Punt JA (2003) Cutting edge: identification of the targets of clonal deletion in an unmanipulated thymus. J Immunol 170:10–13PubMed
41.
Zurück zum Zitat Calnan BJ, Szychowski S, Chan FK, Cado D, Winoto A (1995) A role for the orphan steroid receptor Nur77 in apoptosis accompanying antigen-induced negative selection. Immunity 3:273–282. doi:10.1016/1074-7613(95)90113-2 PubMed Calnan BJ, Szychowski S, Chan FK, Cado D, Winoto A (1995) A role for the orphan steroid receptor Nur77 in apoptosis accompanying antigen-induced negative selection. Immunity 3:273–282. doi:10.​1016/​1074-7613(95)90113-2 PubMed
43.
Zurück zum Zitat Dequiedt F, Van Lint J, Lecomte E, Van Duppen V, Seufferlein T, Vandenheede JR, Wattiez R, Kettmann R (2005) Phosphorylation of histone deacetylase 7 by protein kinase D mediates T cell receptor-induced Nur77 expression and apoptosis. J Exp Med 201:793–804. doi:10.1084/jem.20042034 PubMed Dequiedt F, Van Lint J, Lecomte E, Van Duppen V, Seufferlein T, Vandenheede JR, Wattiez R, Kettmann R (2005) Phosphorylation of histone deacetylase 7 by protein kinase D mediates T cell receptor-induced Nur77 expression and apoptosis. J Exp Med 201:793–804. doi:10.​1084/​jem.​20042034 PubMed
44.
Zurück zum Zitat Parra M, Kasler H, McKinsey TA, Olson EN, Verdin E (2005) Protein kinase D1 phosphorylates HDAC7 and induces its nuclear export after T-cell receptor activation. J Biol Chem 280:13762–13770. doi:10.1074/jbc.M413396200 PubMed Parra M, Kasler H, McKinsey TA, Olson EN, Verdin E (2005) Protein kinase D1 phosphorylates HDAC7 and induces its nuclear export after T-cell receptor activation. J Biol Chem 280:13762–13770. doi:10.​1074/​jbc.​M413396200 PubMed
45.
Zurück zum Zitat Dequiedt F, Kasler H, Fischle W, Kiermer V, Weinstein M, Herndier BG, Verdin E (2003) HDAC7, a thymus-specific class II histone deacetylase, regulates Nur77 transcription and TCR-mediated apoptosis. Immunity 18:687–698. doi:10.1016/S1074-7613(03)00109-2 PubMed Dequiedt F, Kasler H, Fischle W, Kiermer V, Weinstein M, Herndier BG, Verdin E (2003) HDAC7, a thymus-specific class II histone deacetylase, regulates Nur77 transcription and TCR-mediated apoptosis. Immunity 18:687–698. doi:10.​1016/​S1074-7613(03)00109-2 PubMed
47.
Zurück zum Zitat Thompson J, Winoto A (2008) During negative selection, Nur77 family proteins translocate to mitochondria where they associate with Bcl-2 and expose its proapoptotic BH3 domain. J Exp Med 205:1029–1036. doi:10.1084/jem.20080101 PubMed Thompson J, Winoto A (2008) During negative selection, Nur77 family proteins translocate to mitochondria where they associate with Bcl-2 and expose its proapoptotic BH3 domain. J Exp Med 205:1029–1036. doi:10.​1084/​jem.​20080101 PubMed
49.
Zurück zum Zitat Bouillet P, Purton JF, Godfrey DI, Zhang LC, Coultas L, Puthalakath H, Pellegrini M, Cory S, Adams JM, Strasser A (2002) BH3-only Bcl-2 family member Bim is required for apoptosis of autoreactive thymocytes. Nature 415:922–926. doi:10.1038/415922a PubMed Bouillet P, Purton JF, Godfrey DI, Zhang LC, Coultas L, Puthalakath H, Pellegrini M, Cory S, Adams JM, Strasser A (2002) BH3-only Bcl-2 family member Bim is required for apoptosis of autoreactive thymocytes. Nature 415:922–926. doi:10.​1038/​415922a PubMed
50.
Zurück zum Zitat Cante-Barrett K, Gallo EM, Winslow MM, Crabtree GR (2006) Thymocyte negative selection is mediated by protein kinase C- and Ca2+-dependent transcriptional induction of bim. J Immunol 176:2299–2306PubMed Cante-Barrett K, Gallo EM, Winslow MM, Crabtree GR (2006) Thymocyte negative selection is mediated by protein kinase C- and Ca2+-dependent transcriptional induction of bim. J Immunol 176:2299–2306PubMed
51.
Zurück zum Zitat Baldwin TA, Hogquist KA (2007) Transcriptional analysis of clonal deletion in vivo. J Immunol 179:837–844PubMed Baldwin TA, Hogquist KA (2007) Transcriptional analysis of clonal deletion in vivo. J Immunol 179:837–844PubMed
52.
Zurück zum Zitat Liston A, Hardy K, Pittelkow Y, Wilson SR, Makaroff LE, Fahrer AM, Goodnow CC (2007) Impairment of organ-specific T cell negative selection by diabetes susceptibility genes: genomic analysis by mRNA profiling. Genome Biol 8:R12. doi:10.1186/gb-2007-8-1-r12 PubMed Liston A, Hardy K, Pittelkow Y, Wilson SR, Makaroff LE, Fahrer AM, Goodnow CC (2007) Impairment of organ-specific T cell negative selection by diabetes susceptibility genes: genomic analysis by mRNA profiling. Genome Biol 8:R12. doi:10.​1186/​gb-2007-8-1-r12 PubMed
53.
Zurück zum Zitat Liston A, Lesage S, Gray DH, O’Reilly LA, Strasser A, Fahrer AM, Boyd RL, Wilson J, Baxter AG, Gallo EM, Crabtree GR, Peng K, Wilson SR, Goodnow CC (2004) Generalized resistance to thymic deletion in the NOD mouse; a polygenic trait characterized by defective induction of Bim. Immunity 21:817–830PubMed Liston A, Lesage S, Gray DH, O’Reilly LA, Strasser A, Fahrer AM, Boyd RL, Wilson J, Baxter AG, Gallo EM, Crabtree GR, Peng K, Wilson SR, Goodnow CC (2004) Generalized resistance to thymic deletion in the NOD mouse; a polygenic trait characterized by defective induction of Bim. Immunity 21:817–830PubMed
55.
Zurück zum Zitat DeRyckere D, Mann DL, DeGregori J (2003) Characterization of transcriptional regulation during negative selection in vivo. J Immunol 171:802–811PubMed DeRyckere D, Mann DL, DeGregori J (2003) Characterization of transcriptional regulation during negative selection in vivo. J Immunol 171:802–811PubMed
56.
Zurück zum Zitat Bunin A, Khwaja FW, Kersh GJ (2005) Regulation of Bim by TCR signals in CD4/CD8 double-positive thymocytes. J Immunol 175:1532–1539PubMed Bunin A, Khwaja FW, Kersh GJ (2005) Regulation of Bim by TCR signals in CD4/CD8 double-positive thymocytes. J Immunol 175:1532–1539PubMed
57.
Zurück zum Zitat Li QJ, Chau J, Ebert PJ, Sylvester G, Min H, Liu G, Braich R, Manoharan M, Soutschek J, Skare P, Klein LO, Davis MM, Chen CZ (2007) miR-181a is an intrinsic modulator of T cell sensitivity and selection. Cell 129:147–161. doi:10.1016/j.cell.2007.03.008 PubMed Li QJ, Chau J, Ebert PJ, Sylvester G, Min H, Liu G, Braich R, Manoharan M, Soutschek J, Skare P, Klein LO, Davis MM, Chen CZ (2007) miR-181a is an intrinsic modulator of T cell sensitivity and selection. Cell 129:147–161. doi:10.​1016/​j.​cell.​2007.​03.​008 PubMed
58.
Zurück zum Zitat Muljo SA, Ansel KM, Kanellopoulou C, Livingston DM, Rao A, Rajewsky K (2005) Aberrant T cell differentiation in the absence of Dicer. J Exp Med 202:261–269. doi:10.1084/jem.20050678 PubMed Muljo SA, Ansel KM, Kanellopoulou C, Livingston DM, Rao A, Rajewsky K (2005) Aberrant T cell differentiation in the absence of Dicer. J Exp Med 202:261–269. doi:10.​1084/​jem.​20050678 PubMed
59.
Zurück zum Zitat Cobb BS, Hertweck A, Smith J, O’Connor E, Graf D, Cook T, Smale ST, Sakaguchi S, Livesey FJ, Fisher AG, Merkenschlager M (2006) A role for Dicer in immune regulation. J Exp Med 203:2519–2527. doi:10.1084/jem.20061692 PubMed Cobb BS, Hertweck A, Smith J, O’Connor E, Graf D, Cook T, Smale ST, Sakaguchi S, Livesey FJ, Fisher AG, Merkenschlager M (2006) A role for Dicer in immune regulation. J Exp Med 203:2519–2527. doi:10.​1084/​jem.​20061692 PubMed
61.
Zurück zum Zitat Anderson G, Lane PJ, Jenkinson EJ (2007) Generating intrathymic microenvironments to establish T-cell tolerance. Natl Rev 7:954–963 Anderson G, Lane PJ, Jenkinson EJ (2007) Generating intrathymic microenvironments to establish T-cell tolerance. Natl Rev 7:954–963
62.
Zurück zum Zitat von Boehmer H, Hafen K (1986) Minor but not major histocompatibility antigens of thymus epithelium tolerize precursors of cytolytic T cells. Nature 320:626–628. doi:10.1038/320626a0 von Boehmer H, Hafen K (1986) Minor but not major histocompatibility antigens of thymus epithelium tolerize precursors of cytolytic T cells. Nature 320:626–628. doi:10.​1038/​320626a0
63.
Zurück zum Zitat Laufer TM, DeKoning J, Markowitz JS, Lo D, Glimcher LH (1996) Unopposed positive selection and autoreactivity in mice expressing class II MHC only on thymic cortex. Nature 383:81–85. doi:10.1038/383081a0 PubMed Laufer TM, DeKoning J, Markowitz JS, Lo D, Glimcher LH (1996) Unopposed positive selection and autoreactivity in mice expressing class II MHC only on thymic cortex. Nature 383:81–85. doi:10.​1038/​383081a0 PubMed
64.
Zurück zum Zitat Capone M, Romagnoli P, Beermann F, MacDonald HR, van Meerwijk JP (2001) Dissociation of thymic positive and negative selection in transgenic mice expressing major histocompatibility complex class I molecules exclusively on thymic cortical epithelial cells. Blood 97:1336–1342. doi:10.1182/blood.V97.5.1336 PubMed Capone M, Romagnoli P, Beermann F, MacDonald HR, van Meerwijk JP (2001) Dissociation of thymic positive and negative selection in transgenic mice expressing major histocompatibility complex class I molecules exclusively on thymic cortical epithelial cells. Blood 97:1336–1342. doi:10.​1182/​blood.​V97.​5.​1336 PubMed
66.
Zurück zum Zitat Takeuchi M, Iwasaki A, Nomoto K, Yoshikai Y (1992) Rat thymic epithelium positively selects mouse T cells with specificity for rat MHC class II antigens but fails to induce detectable tolerance in the mouse T cells to the rat MHC antigens. Immunobiology 186:421–434PubMed Takeuchi M, Iwasaki A, Nomoto K, Yoshikai Y (1992) Rat thymic epithelium positively selects mouse T cells with specificity for rat MHC class II antigens but fails to induce detectable tolerance in the mouse T cells to the rat MHC antigens. Immunobiology 186:421–434PubMed
67.
Zurück zum Zitat Allison J, Mullbacher A, Cox K, Morahan G, Boyd R, Scollay R, Blanden RV, Miller JF (1990) Selection of the T-cell repertoire in transgenic mice expressing a transplantation antigen in distinct thymus subsets. Proc Biol Sci 241:170–178PubMed Allison J, Mullbacher A, Cox K, Morahan G, Boyd R, Scollay R, Blanden RV, Miller JF (1990) Selection of the T-cell repertoire in transgenic mice expressing a transplantation antigen in distinct thymus subsets. Proc Biol Sci 241:170–178PubMed
69.
Zurück zum Zitat Carlow DA, Teh SJ, Teh HS (1992) Altered thymocyte development resulting from expressing a deleting ligand on selecting thymic epithelium. J Immunol 148:2988–2995PubMed Carlow DA, Teh SJ, Teh HS (1992) Altered thymocyte development resulting from expressing a deleting ligand on selecting thymic epithelium. J Immunol 148:2988–2995PubMed
71.
Zurück zum Zitat Jordan RK, Robinson JH, Hopkinson NA, House KC, Bentley AL (1985) Thymic epithelium and the induction of transplantation tolerance in nude mice. Nature 314:454–456. doi:10.1038/314454a0 PubMed Jordan RK, Robinson JH, Hopkinson NA, House KC, Bentley AL (1985) Thymic epithelium and the induction of transplantation tolerance in nude mice. Nature 314:454–456. doi:10.​1038/​314454a0 PubMed
72.
Zurück zum Zitat Le Douarin NM, Corbel C, Martin C, Coltey M, Salaun J (1989) Induction of tolerance by embryonic thymic epithelial grafts in birds and mammals. Cold Spring Harb Symp Quant Biol 54(Pt 2):777–787PubMed Le Douarin NM, Corbel C, Martin C, Coltey M, Salaun J (1989) Induction of tolerance by embryonic thymic epithelial grafts in birds and mammals. Cold Spring Harb Symp Quant Biol 54(Pt 2):777–787PubMed
73.
Zurück zum Zitat Modigliani Y, Thomas-Vaslin V, Bandeira A, Coltey M, Le Douarin NM, Coutinho A, Salaun J (1995) Lymphocytes selected in allogeneic thymic epithelium mediate dominant tolerance toward tissue grafts of the thymic epithelium haplotype. Proc Natl Acad Sci U S A 92:7555–7559. doi:10.1073/pnas.92.16.7555 PubMed Modigliani Y, Thomas-Vaslin V, Bandeira A, Coltey M, Le Douarin NM, Coutinho A, Salaun J (1995) Lymphocytes selected in allogeneic thymic epithelium mediate dominant tolerance toward tissue grafts of the thymic epithelium haplotype. Proc Natl Acad Sci U S A 92:7555–7559. doi:10.​1073/​pnas.​92.​16.​7555 PubMed
76.
Zurück zum Zitat McCaughtry TM, Baldwin TA, Wilken MS, Hogquist KA (2008) Clonal deletion of thymocytes can occur in the cortex with no involvement of the medulla. J Exp Med (in press) McCaughtry TM, Baldwin TA, Wilken MS, Hogquist KA (2008) Clonal deletion of thymocytes can occur in the cortex with no involvement of the medulla. J Exp Med (in press)
77.
Zurück zum Zitat Murata S, Sasaki K, Kishimoto T, Niwa S, Hayashi H, Takahama Y, Tanaka K (2007) Regulation of CD8+ T cell development by thymus-specific proteasomes. Science (New York, NY) 316:1349–1353 Murata S, Sasaki K, Kishimoto T, Niwa S, Hayashi H, Takahama Y, Tanaka K (2007) Regulation of CD8+ T cell development by thymus-specific proteasomes. Science (New York, NY) 316:1349–1353
78.
Zurück zum Zitat Honey K, Nakagawa T, Peters C, Rudensky A (2002) Cathepsin L regulates CD4+ T cell selection independently of its effect on invariant chain: a role in the generation of positively selecting peptide ligands. J Exp Med 195:1349–1358. doi:10.1084/jem.20011904 PubMed Honey K, Nakagawa T, Peters C, Rudensky A (2002) Cathepsin L regulates CD4+ T cell selection independently of its effect on invariant chain: a role in the generation of positively selecting peptide ligands. J Exp Med 195:1349–1358. doi:10.​1084/​jem.​20011904 PubMed
79.
Zurück zum Zitat Honey K, Rudensky AY (2003) Lysosomal cysteine proteases regulate antigen presentation. Natl Rev 3:472–482 Honey K, Rudensky AY (2003) Lysosomal cysteine proteases regulate antigen presentation. Natl Rev 3:472–482
80.
Zurück zum Zitat Kurobe H, Liu C, Ueno T, Saito F, Ohigashi I, Seach N, Arakaki R, Hayashi Y, Kitagawa T, Lipp M, Boyd RL, Takahama Y (2006) CCR7-dependent cortex-to-medulla migration of positively selected thymocytes is essential for establishing central tolerance. Immunity 24:165–177. doi:10.1016/j.immuni.2005.12.011 PubMed Kurobe H, Liu C, Ueno T, Saito F, Ohigashi I, Seach N, Arakaki R, Hayashi Y, Kitagawa T, Lipp M, Boyd RL, Takahama Y (2006) CCR7-dependent cortex-to-medulla migration of positively selected thymocytes is essential for establishing central tolerance. Immunity 24:165–177. doi:10.​1016/​j.​immuni.​2005.​12.​011 PubMed
81.
Zurück zum Zitat Zhu M, Chin RK, Tumanov AV, Liu X, Fu YX (2007) Lymphotoxin beta receptor is required for the migration and selection of autoreactive T cells in thymic medulla. J Immunol 179:8069–8075PubMed Zhu M, Chin RK, Tumanov AV, Liu X, Fu YX (2007) Lymphotoxin beta receptor is required for the migration and selection of autoreactive T cells in thymic medulla. J Immunol 179:8069–8075PubMed
82.
Zurück zum Zitat Ueno T, Saito F, Gray DH, Kuse S, Hieshima K, Nakano H, Kakiuchi T, Lipp M, Boyd RL, Takahama Y (2004) CCR7 signals are essential for cortex-medulla migration of developing thymocytes. J Exp Med 200:493–505. doi:10.1084/jem.20040643 PubMed Ueno T, Saito F, Gray DH, Kuse S, Hieshima K, Nakano H, Kakiuchi T, Lipp M, Boyd RL, Takahama Y (2004) CCR7 signals are essential for cortex-medulla migration of developing thymocytes. J Exp Med 200:493–505. doi:10.​1084/​jem.​20040643 PubMed
83.
Zurück zum Zitat Kwan J, Killeen N (2004) CCR7 directs the migration of thymocytes into the thymic medulla. J Immunol 172:3999–4007PubMed Kwan J, Killeen N (2004) CCR7 directs the migration of thymocytes into the thymic medulla. J Immunol 172:3999–4007PubMed
84.
Zurück zum Zitat Hogquist KA, Baldwin TA, Jameson SC (2005) Central tolerance: learning self-control in the thymus. Natl Rev 5:772–782CrossRef Hogquist KA, Baldwin TA, Jameson SC (2005) Central tolerance: learning self-control in the thymus. Natl Rev 5:772–782CrossRef
85.
Zurück zum Zitat Kajiura F, Sun S, Nomura T, Izumi K, Ueno T, Bando Y, Kuroda N, Han H, Li Y, Matsushima A, Takahama Y, Sakaguchi S, Mitani T, Matsumoto M (2004) NF-kappa B-inducing kinase establishes self-tolerance in a thymic stroma-dependent manner. J Immunol 172:2067–2075PubMed Kajiura F, Sun S, Nomura T, Izumi K, Ueno T, Bando Y, Kuroda N, Han H, Li Y, Matsushima A, Takahama Y, Sakaguchi S, Mitani T, Matsumoto M (2004) NF-kappa B-inducing kinase establishes self-tolerance in a thymic stroma-dependent manner. J Immunol 172:2067–2075PubMed
86.
Zurück zum Zitat Akiyama T, Maeda S, Yamane S, Ogino K, Kasai M, Kajiura F, Matsumoto M, Inoue J (2005) Dependence of self-tolerance on TRAF6-directed development of thymic stroma. Science (New York, NY) 308:248–251 Akiyama T, Maeda S, Yamane S, Ogino K, Kasai M, Kajiura F, Matsumoto M, Inoue J (2005) Dependence of self-tolerance on TRAF6-directed development of thymic stroma. Science (New York, NY) 308:248–251
87.
Zurück zum Zitat Nakagawa K, Iwabuchi K, Ogasawara K, Ato M, Kajiwara M, Nishihori H, Iwabuchi C, Ishikura H, Good RA, Onoe K (1997) Generation of NK1.1+ T cell antigen receptor alpha/beta+ thymocytes associated with intact thymic structure. Proc Natl Acad Sci U S A 94:2472–2477. doi:10.1073/pnas.94.6.2472 PubMed Nakagawa K, Iwabuchi K, Ogasawara K, Ato M, Kajiwara M, Nishihori H, Iwabuchi C, Ishikura H, Good RA, Onoe K (1997) Generation of NK1.1+ T cell antigen receptor alpha/beta+ thymocytes associated with intact thymic structure. Proc Natl Acad Sci U S A 94:2472–2477. doi:10.​1073/​pnas.​94.​6.​2472 PubMed
88.
Zurück zum Zitat Bensinger SJ, Bandeira A, Jordan MS, Caton AJ, Laufer TM (2001) Major histocompatibility complex class II-positive cortical epithelium mediates the selection of CD4(+)25(+) immunoregulatory T cells. J Exp Med 194:427–438. doi:10.1084/jem.194.4.427 PubMed Bensinger SJ, Bandeira A, Jordan MS, Caton AJ, Laufer TM (2001) Major histocompatibility complex class II-positive cortical epithelium mediates the selection of CD4(+)25(+) immunoregulatory T cells. J Exp Med 194:427–438. doi:10.​1084/​jem.​194.​4.​427 PubMed
89.
Zurück zum Zitat Eck SC, Zhu P, Pepper M, Bensinger SJ, Freedman BD, Laufer TM (2006) Developmental alterations in thymocyte sensitivity are actively regulated by MHC class II expression in the thymic medulla. J Immunol 176:2229–2237PubMed Eck SC, Zhu P, Pepper M, Bensinger SJ, Freedman BD, Laufer TM (2006) Developmental alterations in thymocyte sensitivity are actively regulated by MHC class II expression in the thymic medulla. J Immunol 176:2229–2237PubMed
90.
Zurück zum Zitat Davalos-Misslitz AC, Worbs T, Willenzon S, Bernhardt G, Forster R (2007) Impaired responsiveness to T-cell receptor stimulation and defective negative selection of thymocytes in CCR7-deficient mice. Blood 110:4351–4359. doi:10.1182/blood-2007-01-070284 PubMed Davalos-Misslitz AC, Worbs T, Willenzon S, Bernhardt G, Forster R (2007) Impaired responsiveness to T-cell receptor stimulation and defective negative selection of thymocytes in CCR7-deficient mice. Blood 110:4351–4359. doi:10.​1182/​blood-2007-01-070284 PubMed
92.
Zurück zum Zitat Anderson MS, Venanzi ES, Klein L, Chen Z, Berzins SP, Turley SJ, von Boehmer H, Bronson R, Dierich A, Benoist C, Mathis D (2002) Projection of an immunological self shadow within the thymus by the aire protein. Science (New York, NY) 298:1395–1401 Anderson MS, Venanzi ES, Klein L, Chen Z, Berzins SP, Turley SJ, von Boehmer H, Bronson R, Dierich A, Benoist C, Mathis D (2002) Projection of an immunological self shadow within the thymus by the aire protein. Science (New York, NY) 298:1395–1401
93.
Zurück zum Zitat Liston A, Lesage S, Wilson J, Peltonen L, Goodnow CC (2003) Aire regulates negative selection of organ-specific T cells. Nat Immunol 4:350–354. doi:10.1038/ni906 PubMed Liston A, Lesage S, Wilson J, Peltonen L, Goodnow CC (2003) Aire regulates negative selection of organ-specific T cells. Nat Immunol 4:350–354. doi:10.​1038/​ni906 PubMed
95.
Zurück zum Zitat White AJ, Withers DR, Parnell SM, Scott HS, Finke D, Lane PJ, Jenkinson EJ, Anderson G (2008) Sequential phases in the development of Aire-expressing medullary thymic epithelial cells involve distinct cellular input. Eur J Immunol 38:942–947. doi:10.1002/eji.200738052 PubMed White AJ, Withers DR, Parnell SM, Scott HS, Finke D, Lane PJ, Jenkinson EJ, Anderson G (2008) Sequential phases in the development of Aire-expressing medullary thymic epithelial cells involve distinct cellular input. Eur J Immunol 38:942–947. doi:10.​1002/​eji.​200738052 PubMed
96.
Zurück zum Zitat Rossi SW, Kim MY, Leibbrandt A, Parnell SM, Jenkinson WE, Glanville SH, McConnell FM, Scott HS, Penninger JM, Jenkinson EJ, Lane PJ, Anderson G (2007) RANK signals from CD4(+)3(−) inducer cells regulate development of Aire-expressing epithelial cells in the thymic medulla. J Exp Med 204:1267–1272. doi:10.1084/jem.20062497 PubMed Rossi SW, Kim MY, Leibbrandt A, Parnell SM, Jenkinson WE, Glanville SH, McConnell FM, Scott HS, Penninger JM, Jenkinson EJ, Lane PJ, Anderson G (2007) RANK signals from CD4(+)3(−) inducer cells regulate development of Aire-expressing epithelial cells in the thymic medulla. J Exp Med 204:1267–1272. doi:10.​1084/​jem.​20062497 PubMed
97.
Zurück zum Zitat Boehm T, Scheu S, Pfeffer K, Bleul CC (2003) Thymic medullary epithelial cell differentiation, thymocyte emigration, and the control of autoimmunity require lympho-epithelial cross talk via LTbetaR. J Exp Med 198:757–769. doi:10.1084/jem.20030794 PubMed Boehm T, Scheu S, Pfeffer K, Bleul CC (2003) Thymic medullary epithelial cell differentiation, thymocyte emigration, and the control of autoimmunity require lympho-epithelial cross talk via LTbetaR. J Exp Med 198:757–769. doi:10.​1084/​jem.​20030794 PubMed
98.
Zurück zum Zitat Burkly L, Hession C, Ogata L, Reilly C, Marconi LA, Olson D, Tizard R, Cate R, Lo D (1995) Expression of relB is required for the development of thymic medulla and dendritic cells. Nature 373:531–536. doi:10.1038/373531a0 PubMed Burkly L, Hession C, Ogata L, Reilly C, Marconi LA, Olson D, Tizard R, Cate R, Lo D (1995) Expression of relB is required for the development of thymic medulla and dendritic cells. Nature 373:531–536. doi:10.​1038/​373531a0 PubMed
99.
Zurück zum Zitat Kinoshita D, Hirota F, Kaisho T, Kasai M, Izumi K, Bando Y, Mouri Y, Matsushima A, Niki S, Han H, Oshikawa K, Kuroda N, Maegawa M, Irahara M, Takeda K, Akira S, Matsumoto M (2006) Essential role of IkappaB kinase alpha in thymic organogenesis required for the establishment of self-tolerance. J Immunol 176:3995–4002PubMed Kinoshita D, Hirota F, Kaisho T, Kasai M, Izumi K, Bando Y, Mouri Y, Matsushima A, Niki S, Han H, Oshikawa K, Kuroda N, Maegawa M, Irahara M, Takeda K, Akira S, Matsumoto M (2006) Essential role of IkappaB kinase alpha in thymic organogenesis required for the establishment of self-tolerance. J Immunol 176:3995–4002PubMed
100.
Zurück zum Zitat Gabler J, Arnold J, Kyewski B (2007) Promiscuous gene expression and the developmental dynamics of medullary thymic epithelial cells. Eur J Immunol 37:3363–3372. doi:10.1002/eji.200737131 PubMed Gabler J, Arnold J, Kyewski B (2007) Promiscuous gene expression and the developmental dynamics of medullary thymic epithelial cells. Eur J Immunol 37:3363–3372. doi:10.​1002/​eji.​200737131 PubMed
101.
Zurück zum Zitat Gillard GO, Farr AG (2006) Features of medullary thymic epithelium implicate postnatal development in maintaining epithelial heterogeneity and tissue-restricted antigen expression. J Immunol 176:5815–5824PubMed Gillard GO, Farr AG (2006) Features of medullary thymic epithelium implicate postnatal development in maintaining epithelial heterogeneity and tissue-restricted antigen expression. J Immunol 176:5815–5824PubMed
102.
Zurück zum Zitat Hubert FX, Kinkel SA, Webster KE, Cannon P, Crewther PE, Proeitto AI, Wu L, Heath WR, Scott HS (2008) A specific anti-Aire antibody reveals aire expression is restricted to medullary thymic epithelial cells and not expressed in periphery. J Immunol 180:3824–3832PubMed Hubert FX, Kinkel SA, Webster KE, Cannon P, Crewther PE, Proeitto AI, Wu L, Heath WR, Scott HS (2008) A specific anti-Aire antibody reveals aire expression is restricted to medullary thymic epithelial cells and not expressed in periphery. J Immunol 180:3824–3832PubMed
103.
Zurück zum Zitat Gray D, Abramson J, Benoist C, Mathis D (2007) Proliferative arrest and rapid turnover of thymic epithelial cells expressing Aire. J Exp Med 204:2521–2528. doi:10.1084/jem.20070795 PubMed Gray D, Abramson J, Benoist C, Mathis D (2007) Proliferative arrest and rapid turnover of thymic epithelial cells expressing Aire. J Exp Med 204:2521–2528. doi:10.​1084/​jem.​20070795 PubMed
104.
Zurück zum Zitat Johnnidis JB, Venanzi ES, Taxman DJ, Ting JP, Benoist CO, Mathis DJ (2005) Chromosomal clustering of genes controlled by the aire transcription factor. Proc Natl Acad Sci U S A 102:7233–7238. doi:10.1073/pnas.0502670102 PubMed Johnnidis JB, Venanzi ES, Taxman DJ, Ting JP, Benoist CO, Mathis DJ (2005) Chromosomal clustering of genes controlled by the aire transcription factor. Proc Natl Acad Sci U S A 102:7233–7238. doi:10.​1073/​pnas.​0502670102 PubMed
105.
Zurück zum Zitat Derbinski J, Pinto S, Rosch S, Hexel K, Kyewski B (2008) Promiscuous gene expression patterns in single medullary thymic epithelial cells argue for a stochastic mechanism. Proc Natl Acad Sci U S A 105:657–662. doi:10.1073/pnas.0707486105 PubMed Derbinski J, Pinto S, Rosch S, Hexel K, Kyewski B (2008) Promiscuous gene expression patterns in single medullary thymic epithelial cells argue for a stochastic mechanism. Proc Natl Acad Sci U S A 105:657–662. doi:10.​1073/​pnas.​0707486105 PubMed
106.
Zurück zum Zitat Derbinski J, Schulte A, Kyewski B, Klein L (2001) Promiscuous gene expression in medullary thymic epithelial cells mirrors the peripheral self. Nat Immunol 2:1032–1039. doi:10.1038/ni723 PubMed Derbinski J, Schulte A, Kyewski B, Klein L (2001) Promiscuous gene expression in medullary thymic epithelial cells mirrors the peripheral self. Nat Immunol 2:1032–1039. doi:10.​1038/​ni723 PubMed
107.
Zurück zum Zitat DeVoss J, Hou Y, Johannes K, Lu W, Liou GI, Rinn J, Chang H, Caspi RR, Fong L, Anderson MS (2006) Spontaneous autoimmunity prevented by thymic expression of a single self-antigen. J Exp Med 203:2727–2735. doi:10.1084/jem.20061864 PubMed DeVoss J, Hou Y, Johannes K, Lu W, Liou GI, Rinn J, Chang H, Caspi RR, Fong L, Anderson MS (2006) Spontaneous autoimmunity prevented by thymic expression of a single self-antigen. J Exp Med 203:2727–2735. doi:10.​1084/​jem.​20061864 PubMed
110.
Zurück zum Zitat Seach N, Ueno T, Fletcher AL, Lowen T, Mattesich M, Engwerda CR, Scott HS, Ware CF, Chidgey AP, Gray DH, Boyd RL (2008) The lymphotoxin pathway regulates Aire-independent expression of ectopic genes and chemokines in thymic stromal cells. J Immunol 180:5384–5392PubMed Seach N, Ueno T, Fletcher AL, Lowen T, Mattesich M, Engwerda CR, Scott HS, Ware CF, Chidgey AP, Gray DH, Boyd RL (2008) The lymphotoxin pathway regulates Aire-independent expression of ectopic genes and chemokines in thymic stromal cells. J Immunol 180:5384–5392PubMed
111.
Zurück zum Zitat Viret C, Sant’Angelo DB, He X, Ramaswamy H, Janeway CA Jr (2001) A role for accessibility to self-peptide-self-MHC complexes in intrathymic negative selection. J Immunol 166:4429–4437PubMed Viret C, Sant’Angelo DB, He X, Ramaswamy H, Janeway CA Jr (2001) A role for accessibility to self-peptide-self-MHC complexes in intrathymic negative selection. J Immunol 166:4429–4437PubMed
112.
Zurück zum Zitat Brocker T (1999) The role of dendritic cells in T cell selection and survival. J Leukoc Biol 66:331–335PubMed Brocker T (1999) The role of dendritic cells in T cell selection and survival. J Leukoc Biol 66:331–335PubMed
113.
Zurück zum Zitat Donskoy E, Goldschneider I (2003) Two developmentally distinct populations of dendritic cells inhabit the adult mouse thymus: demonstration by differential importation of hematogenous precursors under steady state conditions. J Immunol 170:3514–3521PubMed Donskoy E, Goldschneider I (2003) Two developmentally distinct populations of dendritic cells inhabit the adult mouse thymus: demonstration by differential importation of hematogenous precursors under steady state conditions. J Immunol 170:3514–3521PubMed
115.
Zurück zum Zitat Watanabe N, Wang YH, Lee HK, Ito T, Wang YH, Cao W, Liu YJ (2005) Hassall’s corpuscles instruct dendritic cells to induce CD4+CD25+ regulatory T cells in human thymus. Nature 436:1181–1185. doi:10.1038/nature03886 PubMed Watanabe N, Wang YH, Lee HK, Ito T, Wang YH, Cao W, Liu YJ (2005) Hassall’s corpuscles instruct dendritic cells to induce CD4+CD25+ regulatory T cells in human thymus. Nature 436:1181–1185. doi:10.​1038/​nature03886 PubMed
116.
Zurück zum Zitat Bonasio R, Scimone ML, Schaerli P, Grabie N, Lichtman AH, von Andrian UH (2006) Clonal deletion of thymocytes by circulating dendritic cells homing to the thymus. Nat Immunol 7:1092–1100. doi:10.1038/ni1385 PubMed Bonasio R, Scimone ML, Schaerli P, Grabie N, Lichtman AH, von Andrian UH (2006) Clonal deletion of thymocytes by circulating dendritic cells homing to the thymus. Nat Immunol 7:1092–1100. doi:10.​1038/​ni1385 PubMed
118.
Zurück zum Zitat Ladi E, Schwickert T, Chtanova T, Chen Y, Yin X, Le Borgne M, Aaron H, Herzmark P, Chan SW, Killeen N, Roysam B, Lipp M, Robey EA (2008) Thymocyte-dendritic cell interactions near sources of CCR7 ligands in the thymic cortex. FASEB J 22:661–669 Ladi E, Schwickert T, Chtanova T, Chen Y, Yin X, Le Borgne M, Aaron H, Herzmark P, Chan SW, Killeen N, Roysam B, Lipp M, Robey EA (2008) Thymocyte-dendritic cell interactions near sources of CCR7 ligands in the thymic cortex. FASEB J 22:661–669
119.
Zurück zum Zitat Buhlmann JE, Elkin SK, Sharpe AH (2003) A role for the B7-1/B7-2:CD28/CTLA-4 pathway during negative selection. J Immunol 170:5421–5428PubMed Buhlmann JE, Elkin SK, Sharpe AH (2003) A role for the B7-1/B7-2:CD28/CTLA-4 pathway during negative selection. J Immunol 170:5421–5428PubMed
Metadaten
Titel
Central tolerance: what have we learned from mice?
verfasst von
Tom M. McCaughtry
Kristin A. Hogquist
Publikationsdatum
01.12.2008
Verlag
Springer-Verlag
Erschienen in
Seminars in Immunopathology / Ausgabe 4/2008
Print ISSN: 1863-2297
Elektronische ISSN: 1863-2300
DOI
https://doi.org/10.1007/s00281-008-0137-0

Weitere Artikel der Ausgabe 4/2008

Seminars in Immunopathology 4/2008 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.