Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Nature Reviews Immunology | AOP, published online 16 May 2014; doi:10.

1038/nri3667 REVIEWS

Positive and negative selection of the


T cell repertoire: what thymocytes see
(and don’t see)
Ludger Klein1, Bruno Kyewski2, Paul M. Allen3 and Kristin A. Hogquist4
Abstract | The fate of developing T cells is specified by the interaction of their antigen
receptors with self-peptide–MHC complexes that are displayed by thymic
antigen-presenting cells (APCs). Various subsets of thymic APCs are strategically positioned
in particular thymic microenvironments and they coordinate the selection of a functional
and self-tolerant T cell repertoire. In this Review, we discuss the different strategies that
these APCs use to sample and process self antigens and to thereby generate partly unique,
‘idiosyncratic’ peptide–MHC ligandomes. We discuss how the particular composition of the
peptide–MHC ligandomes that are presented by specific APC subsets not only shapes the
T cell repertoire in the thymus but may also indelibly imprint the behaviour of mature T cells
in the periphery.

The recognition of self peptides that are embedded in Antigen presentation in the cortex
MHC molecules on thymic antigen-presenting cells At the peak of its productivity, the mouse thymus gener-
(APCs) is crucial for determining the fate of develop- ates around 50 million CD4+CD8+ double-positive (DP)
ing αβ T cells. Somewhat paradoxically, recognition thymocytes each day that audition for selection1. More
of self can elicit diametrically opposed outcomes. On than 90% of these precursors are subject to death by
one hand, it is essential for thymocyte survival and neglect, as they express ‘useless’ T cell receptors (TCRs)
commitment to either the CD4+ or CD8+ T cell line- that do not mediate positive selection. Positive selec-
age (that is, for the positive selection of thymocytes). tion of ‘mainstream’ αβ T cells is contingent on permis-
On the other hand, recognition of self can be a death sive interactions with a single APC type — namely, the
1
Institute for Immunology, verdict for thymocytes, mediating the negative selec- cortical thymic epithelial cells (cTECs). For conceptual
Ludwig Maximilians tion (also known as clonal deletion) of these cells, or clarity, we therefore restrict a more detailed discus-
University, 80336 Munich,
it can skew cells to alternative fates, such as regula- sion of antigen presentation in the cortex to cTECs and
Germany.
2
Division of Developmental tory T (TReg) cell differentiation. The classical affin- their role in positive selection, and only briefly discuss
Immunology, German Cancer ity model of thymocyte selection offers an attractive negative selection in the cortex.
Research Center, 69120 conceptual framework to resolve this apparent contra-
Heidelberg, Germany. diction (BOX 1). However, it does not take into account Cortical thymic epithelial cells
3
Department of Pathology
and Immunology, Washington
the fact that positive selection and negative selection cTECs are arranged in a three-dimensional scaf-
University School of Medicine, mainly occur in discrete thymic micro­environments fold that supports intimate interactions with double-
St. Louis, Missouri 63110, — namely, the cortex and the medulla, respectively. negative (DN) and DP thymocytes. In addition, indi-
USA. Both compartments contain selection niches that are vidual cTECs can form multicellular complexes that
4
Department of Laboratory
composed of different types of APCs (FIG. 1), thereby encompass up to 20 thymocytes, and these cTECs are
Medicine and Pathology,
University of Minnesota, providing microenvironments that coordinate a spa- referred to as thymic nurse cells (TNCs). TNC num-
Minneapolis, Minnesota tial and temporal segregation of thymocyte selection. bers are decreased in TCR-transgenic mice, possibly
55414, USA. In this Review, we focus on recent advances in our as a consequence of the ‘facilitated’ transit of thymo-
Correspondence to L.K. understanding of the key features of individual thymic cytes through β-selection and positive selection2. Thus,
e-mail: ludger.klein@med.uni-
muenchen.de
APC subsets and discuss how these relate to the gen- it seems that TNC formation is not essential for T cell
doi:10.1038/nri3667 eration of a functional and self-tolerant αβ T cell development per se, but that it may result from lengthy
Published online 16 May 2014 repertoire. ‘audition’ events that occur when only a small subset

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 1

© 2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 1 | The affinity model of thymocyte selection


Positive selection
The process by which The affinity model of thymocyte selection (see figure) centres on the strength of the interaction of the T cell receptor
immature double-positive (TCR) with self-peptide–MHC complexes as a crucial determinant of cell fate. Weak interactions are required to
thymocytes that express T cell protect thymocytes from death by neglect and to promote the positive selection of naive T cells. Strong interactions
receptors with intermediate cause negative selection by apoptosis. Studies in TCR-transgenic mice have shown that negatively selecting peptides
affinity and/or avidity for are primarily high-affinity agonists (including cognate ligands; that is, peptides that lead to the activation of mature
self-peptide–MHC complexes
T cells), whereas positively selecting peptides are often low-affinity antagonists or weak agonists (often called altered
are induced to differentiate
into mature single-positive
peptide ligands). Interestingly, in TCR-transgenic systems, high-affinity agonists were also shown to cause clonal
thymocytes. deviation — that is, the redirection of autoreactive T cells into the regulatory T (TReg) cell lineage. A surprisingly broad
range of affinities seems to be permissive for TReg cell differentiation, and as the affinity of the TCR–MHC interaction
Negative selection increases, the propensity to generate TReg cells increases. Thus, a modified version of the affinity model, where TReg cell
(Also known as clonal deletion). differentiation occurs optimally within a window between positive selection and negative selection, is currently
The intrathymic elimination of favoured. Of note, the demarcation between clonal deletion and clonal deviation is remarkably plastic and seems to
double-positive or single- be subject to stochastic influences. For example, in TCR-transgenic systems, intrathymic expression of an agonist
positive thymocytes that ligand can concomitantly promote either TReg cell
express T cell receptors with
or naive T cell fates in some cases, or both TReg cell
high affinity for self antigens. Negative
differentiation and thymocyte deletion in other
cases. Not surprisingly, therefore, the natural selection
Death by neglect TReg cell
A form of programmed cell repertoires of TCRs that are expressed by naive
Death Naive T cell
death that occurs when CD4+ T cells and TReg cells show some overlap. by neglect
double-positive thymocytes Mathematical modelling suggests that distinct

Affinity
fail to engage in positively fates can arise from the interaction between one
selecting interactions with receptor and the same ligand by iterative summing
self-peptide–MHC complexes of TCR signals during multiple interactions, or by
on cortical thymic epithelial Stochastic overlap
changes in thymocyte sensitivity over time
cells within their finite lifespan
of 3–4 days.
(‘developmental tuning’)107.
Nature Reviews | Immunology
β‑selection
The pre‑T cell receptor
(pre-TCR)-driven process by of DP thymocytes meets the positive selection crite- In terms of MHC class II antigen presentation, cTECs
which double-negative ria. Consistent with this, in non-TCR-transgenic mice express the unique lysosomal proteases cathepsin L (also
thymocytes that carry a TNCs were enriched with thymocytes that had second- known as cathepsin L1) and thymus-specific serine
productively rearranged TCR ary TCRα rearrangements2. Whether such unusual selec- protease (TSSP). Deficiency of these proteases results
β‑chain undergo proliferative
expansion and developmental
tion niches are indeed required to promote thymo­cyte in the impaired selection of CD4+ T cells6,7. Cathepsin
progression. survival and/or continued TCR rearrangements remains L-deficient mice show a strongly decreased polyclonal
to be shown. CD4+ T cell repertoire in the thymus6, whereas TSSP-
Peptide–MHC ligandome Why is positive selection crucially dependent on a deficient mice have normal polyclonal CD4+ T cell
(pMHC ligandome). The
single stromal cell type, when tolerance (as discussed numbers but display defective positive selection of
repertoire of peptides that are
bound by MHC molecules. further below) can be mediated by a variety of cell certain MHC class II‑restricted transgenic TCRs, as
types? One might assume that the essential function well as altered antigen-specific CD4+ T cell responses7.
Proteasome of cTECs simply depends on their location and abun- Moreover, cTECs display an unusually high rate of consti-
The standard proteasome is dant surface expression of MHC molecules. However, tutive macroautophagy, a mechanism that can support the
composed of 14 α-subunits
and 14 β-subunits. Three of the
this is not the case. Instead, it is becoming increasingly ‘unconventional’ loading of peptides onto MHC class II
β-subunits (β1, β2 and β5) are clear that the crucial role of cTECs is, at least in part, a molecules via an endogenous route8. Positive selection
involved in peptide-bond result of the unique machineries that these cells use to of several MHC class II‑restricted transgenic TCRs was
cleavage. Interferon‑γ induces process antigens. It is likely that these proteolytic path- altered upon interference with macroautophagy in the
the expression of the
ways (FIG. 2) — which are reviewed in REF. 3 — endow thymic epithelium, which is consistent with the idea that
immunosubunits β1i, β2i
and β5i that can replace cTECs with a largely unique peptide–MHC ligandome autophagy shapes the MHC class II ligandome of cTECs9.
the catalytic subunits of the (pMHC ligandome) that is distinct from that displayed Bearing in mind that the affinity model of thymocyte
standard proteasome to by any other thymic or peripheral APC. selection does not envisage any need for unique posi-
generate the tively selecting peptides, why may these distinct process-
immunoproteasome, which
has distinct cleavage-site
Antigen processing in cTECs. In terms of MHC class I ing pathways have evolved? Do they generate ‘private’
preferences. antigen presentation, cTECs express a unique cata- peptides that are exclusively displayed by cTECs and
lytic subunit of the proteasome that is referred to as that have unique properties that are required for posi-
Macroautophagy β5t (also known as PSMB11). Proteasomes that incor- tive selection? Or do these peptides simply dilute ubiq-
The generally nonspecific
porate β5t are referred to as ‘thymoproteasomes’. They uitous ‘public’ peptides, which are nonetheless the major
sequestration of cytoplasm
into a double- or multiple- have a substrate preference that is distinct from protea- mediators of positive selection? Alternatively, do pep-
membrane-delimited somes containing the β5 (also known as PSMB5) or β5i tides on cTECs merely have to be different from those
compartment (also known as PSMB8) subunits4, which are termed presented by other thymic APCs? The latter proposition
(autophagosome) of ‘housekeeping proteasomes’ and ‘immunoprotea­ is supported by the finding that the reconstitution of
non-lysosomal origin. Certain
proteins, organelles and
somes’, respectively. Mice that lack thymoproteasomes cathepsin L-deficient mice with MHC class II‑deficient
pathogens may be selectively show a substantial defect in the positive selection of bone marrow — which abrogates the negative selec-
degraded by this process. CD8+ T cells5. tion of CD4+ T cells by haematopoietic APCs — mostly

2 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/immunol

© 2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

a b
Thymocyte Cortex cTEC
• Unique ‘private’ proteolytic pathways
cTEC • Efficient endogenous MHC class II loading
• Constitutive macroautophagy
DN2 • Thymic nurse cell formation

mTEC
Thymic • ‘Promiscuous gene expression’ (AIRE)
DN3
nurse cell • ‘Public’ proteolytic pathways
• Efficient endogenous MHC class II loading
• Macroautophagy
DN1 Migratory cDC
DP • Steady-state immigration from peripheral
sites
• Import of peripheral antigens
• ‘Public’ proteolytic pathways
Blood Migratory • Conventional MHC class II loading
vessel cDC SIRPα+CD8– • Presentation of mTEC-derived and
serum-borne antigens
SP Medulla Resident cDC
Resident cDC
• Intrathymic differentiation
• ‘Public’ proteolytic pathways
mTEC Blood
• Conventional MHC class II loading
vessel
• Presentation of mTEC-derived and
SIRPα–CD8+ serum-borne antigens
Migratory
pDC
mTEC cDC
• Steady-state immigration from peripheral
sites
• Import of peripheral antigens
B cell pDC • ‘Public’ proteolytic pathways
• Conventional MHC class II loading
• No presentation of mTEC-derived TRAs?
B cell
pDC
• Intrathymic or extrathymic origin?
Resident cDC • Efficient presentation of BCR-captured
antigens
• ‘Public’ proteolytic pathways
• No presentation of mTEC-derived TRAs?

Figure 1 | Stromal cell interactions during T cell development.  a | Successive stages of double-negative (DN) T cell
development are accompanied by an outward movement of thymocytes towards the subcapsular Nature Reviews
zone. | Immunology
Subsequent to
β‑selection at the DN3 stage, double-positive (DP) cells ‘randomly walk’ through the outer cortex, which possibly
facilitates the ‘scanning’ of cortical thymic epithelial cells (cTECs) for positively selecting ligands. At this stage, DP
thymocytes may be engulfed by cTECs and form so‑called thymic nurse cells; however, the molecular control and
physiological relevance of this process remains to be established. Interactions between DP cells and cortical
conventional dendritic cells (cDCs) may lead to negative selection. It remains unknown whether these cortical cDCs
exclusively belong to the migratory signal-regulatory protein‑α (SIRPα)-expressing subset. Positively selected CD4 or
CD8 lineage-committed thymocytes relocate into the medulla by directed migration. Upon reaching the medulla,
single-positive (SP) cells again assume a ‘random walk’ motion pattern. Through this random migration, SP cells may now
‘scan’ resident and migratory cDCs, plasmacytoid dendritic cells (pDCs), medullary thymic epithelial cells (mTECs) and
B cells. It is estimated that SP cells engage in around five contacts with antigen-presenting cells (APCs) per hour, so that
during their 4- to 5‑day residency in the medulla, T cells may serially interact with several hundred APCs. Solid arrows
indicate main migratory pathways that are involved in thymocyte selection. Dashed arrows indicate other relevant
migratory pathways. b | Key functional properties of thymic APCs that are discussed in this Review. AIRE, autoimmune
regulator; BCR, B cell receptor; TRA, tissue-restricted antigen.

rescued their CD4+ T cell compartment10. This indicates Nevertheless, several observations concerning the
that the positive selection of CD4+ T cells by cathepsin L‑ role of the thymoproteasome in the selection of CD8+
deficient cTECs is not inefficient per se; however, an T cells suggest a different scenario. Neither the recon-
unusually large fraction of cells that are selected in this stitution with MHC class  I‑deficient bone marrow
way are subject to negative selection. Hence, positive cells nor the inactivation of the gene that encodes
selection on different (but not functionally unique) BCL‑2‑interacting mediator of cell death (BIM; also known
ligands might be necessary to prevent a disproportionate as BCL2L11) rescued the CD8+ T cell compartment of
BCL‑2‑interacting mediator loss of T cells owing to subsequent re‑encounter of the thymo­proteasome-deficient mice11,12. Therefore, the role
of cell death
(BIM). A pro-apoptotic
very same peptides that mediated positive selection in of thymoproteasome-dependent peptides cannot be to
molecule that is crucial for a ‘negatively selecting setting’ — namely, on medullary avoid excessive thymocyte deletion. Gene-replacement
negative selection. APCs that express abundant co‑stimulatory molecules3. experiments provide further evidence for the notion that

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 3

© 2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

‘Private’ peptide ‘Private’ peptide ‘Public’ peptide ‘Public’ peptide


Exogenous
MHC class I MHC class II protein

cTEC only DC, mTEC


or cTEC

Exogenous pathway
very inefficient
Major pathway in DCs
ER but inefficient in mTECs

Cathepsin L Lysosome
and TSSP? or MIIC
TAP Cathepsin S

Minor pathway in DCs


Housekeeping
Thymo- but efficient in mTECs
proteasome (β5)
proteasome and immuno-
(β5t) proteasome (β5i)

Endogenous
protein
Macroautophagosome

Figure 2 | Unique proteolytic pathways generate ‘private’ MHC-bound peptides in cTECs.  Processing of a given
endogenous protein substrate by cortical thymic epithelial cells (cTECs) may give rise toNature
uniqueReviews
‘private’| Immunology
peptides
(left panel), which differ from the ‘public’ peptides (right panel) that are generated by cTECs as well as medullary
thymic epithelial cells (mTECs) and dendritic cells (DCs). MHC class I‑bound peptides on the surface of cTECs can be
processed by proteasomes that contain the catalytic subunit β5t (so-called thymoproteasomes). Owing to a distinct
proteolytic activity of the thymoproteasome, this is likely to lead to the generation of cTEC-specific ‘private’ peptide
epitopes that differ from the ‘public’ epitopes that are generated through the housekeeping proteasome (β5) or the
immunoproteasome (β5i). MHC class II‑bound peptides on cTECs seem to be mostly derived from an unconventional,
endogenous MHC class II‑loading pathway that involves the macroautophagy-mediated shuttling of cytoplasmic
proteins into lysosomes. In this proteolytic compartment, processing by the proteases cathepsin L and
thymus-specific serine protease (TSSP) may generate unique ‘private’ peptides. MHC class II‑bound peptides on
mTECs may likewise be mostly derived from macroautophagy-mediated endogenous MHC class II loading;
however, the lysosomal proteases that generate MHC class II‑bound peptides in mTECs differ from those in cTECs,
being essentially identical to those used by DCs for the processing of exogenously derived substrates along the
‘conventional’ exogenous MHC class II pathway. Of note, it is likely that the peptide–MHC (pMHC) ligandome of
cTECs represents a mixture of ‘private’ and ‘public’ peptides that are uniquely present on cTECs or are shared with
other antigen-presenting cells (APCs), respectively (see FIG. 4). ER, endoplasmic reticulum; MIIC, MHC class II
compartment; TAP, transporter associated with antigen processing.

it is the actual nature of the peptides that are generated by The putative significance of ‘private’ peptides. How
the thymoproteasome — rather than a mere difference could ‘private’ peptides on cTECs be specialized for
between the pMHC repertoires of cTECs and other APCs positive selection? They might bind MHC molecules
— that matters. By inserting the gene that encodes β5i more weakly, as suggested by the observation that
into the locus of the gene that encodes β5t in β5i‑deficient β5t‑containing proteasomes, in contrast to those that
mice, animals were engineered in which, independently of contain β5 or β5i, inefficiently cleave substrates adja-
β5t, the MHC class I ligandomes differed between cTECs cent to hydrophobic amino acids5,13. MHC class I mol-
MHC anchor residues
Amino acid residues of an and other APCs (in this case shaped by the immuno­ ecules preferentially bind peptides with hydrophobic
antigenic peptide that bind in proteasome versus the housekeeping protea­some, respec- C termini. Therefore, ‘wobbly’ binding of β5t‑derived
pockets in the peptide-binding tively)12. This difference alone did not restore positive peptides might result in a faster TCR off-rate and
groove of a major selection in these animals; by inference, peptides that thereby promote positive selection — a scenario sim-
histocompatibility molecule
and account for much of the
are generated by β5t‑containing thymoproteasomes are ilar to the generation of partial agonists by altering
binding energy and specificity not only different but may also somehow bear unique the MHC anchor residues of immunogenic peptides14.
of binding. biophysical features that are related to positive selection. Although attempts to compare the stability of pMHC

4 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/immunol

© 2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

complexes on cTECs with that on other APCs have so CD5low cells nonetheless do constitute a consider-
far failed to disclose such differences11,12, there is inde- able fraction of the steady-state T cell repertoire19. So
pendent evidence that β5t engenders a bias towards why would this be beneficial or even necessary for a
‘weak’ interactions for positive selection. CD5 expres- functional immune system? First, a repertoire that is
sion levels on single-positive (SP) thymocytes are solely composed of clones with high self-reactivity
thought to reflect the signalling intensity of the posi- might be prone to inciting autoimmunity. However,
tively selecting TCR–pMHC interaction, and ‘tuned’ there is as yet no evidence to support this notion. For
CD5 levels persist on mature peripheral T cells as a instance, β5t‑deficient mice display intact negative
footprint of thymic selection15. Intriguingly, the dimin- selection11 and do not exhibit any signs of autoimmun-
ished CD8+ SP thymocyte compartment that is found ity. Second, the presumed competitive disadvantage
in β5t‑deficient mice is mostly composed of cells that of CD5low clones that are selected through low-affin-
express elevated levels of CD5 as well as NR4A1(also ity interactions may in fact not be a general rule.
known as NUR77), which suggests that positive selec- Persaud et al.20 compared two CD4+ T cell clones with
tion in the absence of β5t mostly entails interactions identical affinities for a Listeria monocytogenes anti-
that are of relatively higher affinity 12. In the same vein, gen; one was presumably selected and maintained by
studies in TCR-transgenic mice showed that selec- weak interactions with self, as inferred from its CD5low
tion of ‘natural’ CD5low clones — such as CD8+ T cells phenotype, and the other was selected by stronger
expressing the HY TCR — is highly dependent on β5t, interactions — that is, being CD5 hi and displaying
whereas selection of CD5 hi clones — such as those higher TCRζ phosphorylation. This alleged differential
expressing the OT‑I TCR — is not. However, in studies signalling strength during positive selection correlated
using five different TCR-transgenic mice, the extent with differences in basal TCR signalling in mature
of the β5t dependency of developing thymocytes did peripheral cells. Thus, the CD5hi clone was poised to
not show a perfect inverse correlation with their CD5 produce more IL‑2 and this property, consistent with
expression levels11. Thus, thymoproteasome-derived the ‘tonic signalling’ hypothesis, was actively main-
peptides, and possibly private peptides that are gen- tained in the periphery through the recognition of
erated through other cTEC-specific pathways, might unknown self-peptide–MHC ligands. Regardless of
favour the selection of CD5low T cell clones. this difference, in the course of an L. monocytogenes
infection, CD5low and CD5hi T cells showed compara-
Selection of TCRs across a range of affinities for self. ble proliferative responses, and the CD5hi clone even-
It has been suggested that the re‑exposure of mature tually underwent more extensive cell death, so that in
T cells to their positively selecting peptide (or peptides) this case the CD5low clone ultimately dominated the
CD5
A membrane protein that
is necessary for homeostasis through continual tonic TCR immune response.
associates with the T cell stimulation16. According to this scenario, T cells selected Taken together, unusual antigen processing in cTECs
receptor (TCR) complex. It on ‘private’ pMHC ligands that are not re‑encountered seems to diversify the T cell repertoire for maximal
modulates the TCR signal outside the thymus are predicted to have a competi- versatility, as best exemplified by the thymoprotea-
transduction cascade through
tive disadvantage during steady-state homeostasis. some and CD8+ T cell selection. Interference with this
interactions with various
kinases and phosphatases. Consistent with this idea, mature CD5low T cells in cTEC-specific pathway of pMHC generation results
secondary lymphoid tissues are indeed less responsive in a ‘crippled’ CD8+ T cell repertoire that seems to be
NR4A1 to homeostatic cytokines when compared with their dominated by T cells with a higher affinity for self anti-
NR4A1 encodes an orphan CD5hi counterparts17,18. In further support of such a gens. Corresponding consequences of cathepsin L defi-
nuclear receptor that is
upregulated by T cell receptor
link between thymic pMHC experience and mature ciency or TSSP deficiency for the peripheral CD4+ T cell
signalling in thymocytes and T cell homeostasis, CD5 low T cells that express the repertoire have yet to be described.
mature T cells. β5t‑dependent HY TCR are notoriously poor at homeo­
static proliferation, whereas CD5hi cells expressing the Negative selection in the cortex
HY TCR
OT‑I TCR, which is selected fairly efficiently in the As pointed out already, the vast majority of thymocyte
An MHC class I‑restricted
transgenic T cell receptor (TCR) absence of β5t, show robust homeostatic expansion11. death in the cortex can be attributed to the failure of a
that recognizes a self antigen Also, TCRs of CD5low cells are ‘preloaded’ with less basal large proportion of DP cells to undergo positive selec-
that is encoded by the Y phosphorylation compared with TCRs of CD5hi cells, tion21. Nonetheless, there is also a substantial loss of
chromosome. which might put them at a competitive disadvantage DP thymocytes through negative selection. Recent data
OT-I TCR
in responding to foreign antigens16,19. Indeed, in several show that the number of thymocytes that die through
An MHC class I‑restricted models of infection in which polyclonal CD4+ T cell negative selection in the cortex is in fact much higher
transgenic T cell receptor (TCR) responses to pathogens have been examined, CD5 hi than previously appreciated and may even exceed the
that recognizes a peptide T cells outcompeted CD5low T cells19. This observation number of cells that pass through positive selection22,23.
epitope from ovalbumin.
led to the suggestion that the raison d’être of positive Using a TCR signalling reporter to identify thymocytes
Tonic TCR stimulation selection, rather than imprinting self-MHC restriction, that were rescued from deletion in mice lacking BIM,
Continuous ‘subthreshold’ is to bias T cell selection towards strongly self-reactive it was estimated that 5 × 105 cells undergo negative
recognition of self-peptide– clones that are endowed with a homeostatic advantage selection in the cortex every day 23. This figure not only
MHC complexes by mature and a head start in anti-pathogen responses19. Hence, exceeds the estimated number of positively selected
T cells, which results in a basal
activation state that enables
the idea that private peptides skew positive selection cells but is also around twofold higher than the number
T cells to rapidly respond to towards CD5low T cells that weakly respond to self may of cells that are believed to undergo negative selection
foreign antigen. appear counter-intuitive. in the medulla.

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 5

© 2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

Tissue-restricted antigens Intriguingly, cortical negative selection of thymo- (reviewed in REFS 3,26). Central hallmarks of the thymic
(TRAs). Self constituents cytes that are specific for ‘ubiquitous’ self antigens was medulla that specify its crucial tolerogenic role include
encoded by genes that are shown to depend on a crucial contribution from den- the ‘ectopic’ expression of a myriad of tissue-restricted
expressed by only one or a few dritic cells (DCs). The heterogeneity and functional antigens (TRAs) by medullary thymic epithelial cells
tissue-specific cell lineages as
opposed to housekeeping
attributes of thymic DCs are discussed below in the sec- (mTECs) and the presence of a unique ensemble of
genes. The term TRA is an tion on medullary APCs. At this point, it may suffice to specialized haematopoietic APCs.
operational definition based on highlight that the crucial role of DCs in cortical negative
available expression selection is all the more remarkable considering that Medullary thymic epithelial cells
catalogues, according to which
there are very few DCs in the cortex compared with The phenomenon of promiscuous gene expression in
TRAs are expressed in less
than 5 of the 60 tested tissues.
the medulla and because ‘ubiquitous’ antigens are also mTECs has been reviewed in detail elsewhere27,28. Some
predicted to be displayed by cTECs24. Possibly, these salient features of promiscuous gene expression and
observations reflect an inherent inefficacy of cTECs novel insights are highlighted in BOX 2. Although the
to support negative selection. Consistent with this, entire mTEC population collectively expresses almost
imaging analyses of cortical negative selection in situ all ‘peripheral’ transcripts, each TRA is only expressed
revealed that thymocytes arrest and signal adjacent to by a minor fraction (1–3%) of mTECs at any given time
DCs, even when antigen is also displayed by cTECs25. (FIG. 3). How this mosaic expression pattern ultimately
Because these experiments involved the exogenous translates into the faithful presentation of thousands
delivery of agonist peptide, cTEC-specific pathways of of self antigens in a way that ensures efficient tolerance
antigen processing are unlikely to be the sole determi- remains puzzling.
nant of this impaired capacity of cTECs to induce clonal Self antigens that are expressed by mTECs may be
deletion. Future experimentation is needed to assess seen by T cells in two ways (FIG. 3): first, through ‘auton-
the contribution of other candidate parameters such as omous’ presentation by mTECs themselves or, second,
co‑stimulation, cell adhesion and MHC turnover. through antigen handover and presentation by neigh-
bouring APCs. Direct presentation of endogenously
Antigen presentation in the medulla expressed antigens by mTECs not only induces negative
The medulla serves a crucial function for T cell toler- selection of CD8+ T cells29,30 but also efficiently elicits
ance induction. Indeed, the disruption of the three‑ CD4+ T cell tolerance31–34. At the same time, mTECs
dimensional architecture of the medulla, disturbed are conspicuously inefficient in the ‘conventional’
development of its stromal components, impaired tran- MHC class II‑restricted presentation of extracellular
sit of positively selected thymocytes into the medulla or substrates35,36. Hence, mTECs have apparently evolved
premature egress of thymocytes from the medulla all strategies to bypass the classical exogenous pathways
result in spontaneous manifestations of autoimmunity of MHC class II loading in order to focus their MHC
class II ligandome on endogenous self antigens.
Box 2 | Promiscuous gene expression Endogenous MHC class II loading in mTECs. How do
The term ‘promiscuous gene expression’ refers to the ectopic expression of otherwise mTECs load MHC class II molecules with intracellular
tissue-restricted antigens (TRAs) by medullary thymic epithelial cells (mTECs). antigens? Candidate pathways fall into two categories
Remarkably, besides tissue specificity, promiscuous gene expression violates other (reviewed in REF. 8). The first comprises mechanisms that
fundamental rules of tightly controlled lineage-specific gene expression, such as are dependent on the proteasome and on the transporter
developmental switches and sex specificity, thus comprehensively representing the associated with antigen processing (TAP), which implies
immunological self of peripheral tissues.
a leakage of the content of the endoplasmic reticulum
The autoimmune regulator (AIRE) protein remains the only identified molecular
regulator that is dedicated to controlling a sizeable fraction of the promiscuously
(ER) into MHC class II loading compartments. The sec-
expressed gene pool27. It does not seem to act as a classical sequence-specific ond category comprises processes that are collectively
DNA-binding transcription factor. Instead, AIRE targets and turns on silent gene loci by known as autophagy (‘self eating’): microautophagy,
binding to non-methylated histone 3 lysine 4 (H3K4), which marks promoters that are chaperone-mediated autophagy and macroautophagy.
in closed chromatin regions108,109. It apparently acts as a docking platform for different Their common principle is the delivery of cytoplasmic
multi-protein complexes that are known to facilitate transcription by generating local constituents to lysosomes, which presumably intersects
double-strand breaks, promoting mRNA processing and relieving stalled RNA with the MHC class II loading pathway 37. So far, only
polymerase110,111. Yet none of the presently discussed models of the function of AIRE112 the role of macroautophagy has been examined in the
satisfactorily accounts for all of the intricacies of promiscuous gene expression, most context of thymocyte selection. Athymic nude mice
notably the observation that a given TRA is only expressed by a minor subset (1–3%) of
that were grafted with macroautophagy-deficient thymi
mTECs at any point in time (see FIG. 3). Understanding how this mosaic pattern of
promiscuous gene expression is generated and whether it is maintained in time and
displayed various manifestations of immune-mediated
space at the single-cell level will be crucial for eventually comprehending how central tissue damage, which is consistent with a crucial func-
tolerance can be so efficient, sensitive and stringent. tion of macroautophagy in TECs for loading peptides
Although it was initially thought that promiscuous gene expression in single mTECs is onto MHC class II molecules for T cell repertoire selec-
stochastic52,54, a recent study has revealed distinct and fluctuating patterns of gene tion9. However, these studies did not address whether
co‑expression in subsets of human mTECs, which implies that mTECs transiently display the observed symptoms actually reflected a failure of
TRAs in certain linkage groups53. Co‑expression groups ranged from approximately 100 to negative selection by mTECs or whether they were
300 TRAs, and preferentially mapped to particular chromosomes whereby co‑expressed driven by impaired positive selection by autophagy-
gene loci colocalized within nuclear subdomains, which suggests a level of regulation at deficient cTECs; these two possibilities are not
the higher genome order.
mutually exclusive.

6 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/immunol

© 2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

500 µm

Aire Dmkn Spink3 Clca3

b Antigen ‘handover’
Direct presentation and indirect presentation The medulla as a ‘mosaic’ of self

DC

T cell

TRA-derived
epitope
mTEC

TRA-derived
epitope

Figure 3 | Topological aspects of ‘promiscuous gene expression’ and direct versus indirect presentation of
Nature
tissue-restricted antigens.  a | Any particular tissue-restricted antigen (TRA) is expressed by only Reviews
a small | Immunology
subset of
medullary thymic epithelial cells (mTECs) at any given point in time. In situ hybridization of a section through an entire mouse
thymic lobe for autoimmune regulator (Aire) mRNA expression shows that Aire-expressing cells are densely packed in
medullary regions (M. Aichinger and L.K., unpublished observations). By contrast, transcripts of three representative TRAs —
dermokine (Dmkn), serine protease inhibitor Kazal-type 3 (Spink3) and calcium-activated chloride channel family member 3
(Clca3; also known as Clca1) — are only detectable in very few cells that are scattered throughout medullary areas (dotted
lines). b | Tolerogenic presentation of TRAs that are expressed by few mTECs may occur in two ways that are not mutually
exclusive. The first is through direct presentation by TRA-expressing mTECs themselves, whereby efficient endogenous
MHC class II loading by mTECs in conjunction with serial ‘scanning’ of multiple medullary antigen-presenting cells (APCs)
by thymocytes increases the likelihood of cognate self antigen interactions (left panel). The second is by ‘antigen handover’
to neighbouring conventional dendritic cells (cDCs; middle panel), which may extend the area of tolerogenic presentation
in a mosaic fashion beyond the topologically restricted expression pattern (right panel). The mechanistic details of this
‘directional antigen transfer’ remain to be established. It is conceivable that TRAs are released or shed in a soluble form
to be subsequently captured and processed by cDCs for presentation on MHC class I or class II. Apoptosis of terminally
differentiated mTECs may lead to the release of apoptotic fragments that can also transfer mTEC-derived self antigens to
cDCs. In addition, functional peptide–MHC ligands are unidirectionally translocated from mTECs to DCs42,119.

More recent work has provided compelling evidence with the antigen that was not marked for autophagy 38.
that macroautophagy does indeed support tolerogenic The same study also showed that a mitochondrial ver-
endogenous MHC class II loading in mTECs. When sion of a model antigen required macroautophagy for
two closely related model antigens were targeted to the tolerogenic presentation by TECs, whereas the direct
cytosol of mTECs, a variant that was earmarked for presentation of a membrane-bound form of the same
autophagosomal degradation was presented with much antigen was macroautophagy-independent 38. Possibly,
higher efficacy and displayed a superior capacity to macroautophagy may be generally required for endog-
induce the negative selection of CD4+ T cells compared enous access to MHC class II substrates that reside in the

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 7

© 2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

cytoplasm or within organelles — such as mitochondria, equally possible that the residual MHC class II expres-
peroxisomes or the nucleus —which would be consistent sion on mTECs in C2TAkd mice might still suffice to
with the role of autophagy in sampling these subcellular censor autoreactive CD4+ T cells at the high-affinity end
compartments39. By contrast, membrane proteins seem of the TCR spectrum.
to be inherently able to access MHC class II loading In further support of a substantial autonomous con-
compartments independently of macroautophagy 40. tribution of mTECs as APCs for negative selection of
polyclonal CD4+ T cells, the CD4+ SP thymocyte compart-
Direct versus indirect presentation of self antigens by ment in C2TAkd mice was markedly enlarged31. In fact,
mTECs. A clear delineation of the quantitative or quali- the decreased expression of MHC class II molecules on
tative impact of direct versus indirect presentation of mTECs in these mice and the complete ablation of MHC
TRAs by mTECs or DCs (or any other thymic APC class II expression on DCs in irradiated wild-type mice that
for that matter), respectively, is only slowly emerging, were reconstituted with MHC class II‑deficient bone mar-
partly owing to potential redundancies between the row had a similar effect on the degree of negative selection
two mechanisms. From the study of transgenic neo- within the CD4+ SP thymocyte compartment31. Moreover,
self antigens, there is a wealth of information support- combining haematopoietic MHC class II deficiency with
ing the idea that direct presentation by mTECs is an reduced MHC class II expression on mTECs had an addi-
exquisitely efficient tolerance mechanism (reviewed in tive effect, which suggests a non-redundant contribution
REF. 41). At the same time, there is accruing evidence of both DCs and mTECs to negative selection.
that the medulla provides a specialized microenviron- Given the low frequency of mTECs that express and
ment that is conducive to intercellular antigen trans- hence potentially present a given TRA1, ‘saturating’ toler-
fer 42. However, few experimental models demonstrate ance induction through direct presentation would require
an essential requirement for such antigen handover and exceedingly efficient scanning of numerous mTECs by
some of these findings remain controversial29,43. In a thymocytes (see FIG. 3). Real-time imaging of thymocyte
recent study, MHC class II tetramers were used to mon- motility showed that this is indeed the case: SP thymo-
itor the steady-state negative selection of polyclonal cytes ‘randomly walk’ within medullary areas at a velocity
CD4+ T cells that were reactive to interphotoreceptor of 10 μm per minute, which allows them to engage in mul-
retinoid-binding protein (IRBP; also known as RBP3), tiple contacts with APCs48–50. Estimates of the number of
which is an autoimmune regulator (AIRE)-dependent APCs that can be scanned within the 4- to 5‑day sojourn
TRA that is exclusively expressed by mTECs. Ablating of SP cells in the medulla vary from a few hundred to sev-
MHC class II expression in haematopoietic cells abol- eral thousand1,49,51. Bioinformatic modelling on the basis
ished the negative selection of T cells that are specific of TRA expression data at the single-cell level52–54 predicts
for this physiologically expressed self antigen, which that 200–500 mTECs will be sufficient to cover the full
indicates an essential requirement for inter­cellular TRA repertoire at a given point in time (B.K., H. Mayer
transfer between antigen-expressing mTECs and and S. Pinto, unpublished observations). Shifting TRA
antigen-presenting haematopoietic APCs, at least for expression patterns over time and corresponding fluc-
certain epitopes of IRBP44. tuations in the pMHC ligandome of individual mTECs
A conclusive dissection of the dual role of mTECs would further reduce the minimal number of cells that
(as antigen providers and presenters) in tolerizing need to be scanned, provided that T cells re‑encounter
the polyclonal T cell repertoire remains experimen- the same mTEC over time49,53. Notwithstanding a con-
tally challenging. Selective ablation of either MHC siderable error margin in these calculations, it seems that
class I or MHC class II expression in mTECs by con- T cells may not even need to roam through large volumes
Thymic crosstalk ditional gene targeting has been surprisingly difficult of the medulla in order to saturate TRA encounters that
The mutual developmental to achieve. A further caveat of such an approach is that result from autonomous presentation by mTECs.
dependence of the T cell and MHC class II‑dependent thymic crosstalk between thy-
the stromal cell (that is,
mocytes and mTECs organizes mTEC differentiation45, Thymic dendritic cells
non‑T cell) compartments of
the thymus, which is specified such that the abolition of MHC class II expression on The overall contribution of DCs to total thymic cellu-
by complex receptor–ligand mTECs will most probably affect promiscuous gene larity is in the order of 0.5%. Thymic DCs can be sub­
interactions. expression in qualitative or quantitative terms. To avoid divided into three major subsets55, two of which belong
such confounding effects, an experimental strategy of to the conventional — also known as classical — DC
C2TAkd mice
A mouse strain that expresses
tissue-specific knockdown of MHC class II molecules (cDC) lineage, whereas the remaining subset of thymic
a designer microRNA that in transgenic mice (termed C2TAkd mice ) has been DCs belongs to the plasmacytoid DC (pDC) lineage.
targets the MHC class 2 devised31. The selective attenuation of antigen pres- The heterogeneity of DCs in the thymus raises obvi-
transactivator (C2TA) entation by mTECs in these mice resulted in sporadic ous questions with regard to the possible functional
specifically in medullary thymic
bouts of mild tissue infiltration, yet did not elicit overt specialization of individual subtypes. Determinants of
epithelial cells (mTECs). This
leads to a reduction of MHC autoimmunity. These findings contrast with the sponta- such a division of labour could include cell biological
class II expression to neous autoimmunity that results from AIRE deficiency features of APC function (for example, antigen uptake
approximately 10% of its or even from selectively ablating the expression of single and processing), intrathymic versus extrathymic ori-
physiological levels, while TRAs in mTECs46,47. At first glance, this could be inter- gin and the positioning of DCs within distinct thymic
preserving intact mTEC
differentiation and
preted to indicate that direct antigen presentation to micro­environments. All of these features will eventu-
tissue-restricted antigen CD4+ T cells by mTECs, in contrast to TRA expression, ally define the sampling territories of each subset and
expression. is not essential to prevent autoimmunity; however, it is hence its self-peptide–MHC ligandome.

8 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/immunol

© 2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

Resident versus migratory cDCs. Around two-thirds communication). Notwithstanding these apparent dis-
of thymic DCs can be classified as CD11chiCD45RA− crepancies, there is some consensus that SIRPα+ migra-
cDCs. These can be further subdivided according to dif- tory cDCs sample intravenously injected model antigens
ferential co‑expression of CD8α and signal-regulatory from the bloodstream in vivo more efficiently than
protein‑α (SIRPα; also known as CD172a and SHPS1), resident cDCs35,60–62.
with roughly two-thirds of thymic cDCs displaying a Taken together, the differences between resident and
CD8α+SIRPα− surface phenotype and one-third dis- migratory cDCs with respect to their origin, responsive-
playing a reciprocal CD8α−SIRPα+ surface phenotype55. ness to chemokines and selective occupancy of micro-
The major CD8α+SIRPα− cDC subset originates from environmental niches imply a degree of functional
an intrathymic differentiation pathway, and hence specialization. Resident cDCs preferentially seed the
these cells are commonly referred to as resident cDCs, medulla and seem to be best equipped to focus their
whereas the minor CD8α−SIRPα+ cDC subset is main- antigen display on self antigens that are captured within
tained by steady-state immigration of cells from the the thymic microenvironment. By contrast, migratory
periphery, and these cells are therefore referred to as cDCs are found both in the medulla and the cortex
migratory cDCs56. and may serve a dual role: first, to transport peripher-
Resident cDCs in the thymus bear obvious pheno- ally acquired self antigens into the thymus, and second,
typic resemblance to CD8α+ cDCs in the periphery. to capture and present blood-borne self antigens. It is
Peripheral CD8α+ cDCs are known to be particularly unlikely, however, that there is a strict demarcation
efficient in cross-presentation — that is, the presentation between these functions.
of exogenous antigens in the context of MHC class I57.
Indeed, thymic CD8α+SIRPα− cDCs showed a superior Plasmacytoid dendritic cells. Roughly one-third of
cross-presentation capacity in vitro when compared all thymic DCs are pDCs. pDCs enter the thymus as
to the migratory subset 58. In vivo, intrathymic cross- a migratory population from peripheral sites56, which
presentation was found to contribute to CD8+ T cell tol- indicates a close lineage relationship between periph-
erance towards a model antigen mimicking a TRA-like eral and thymic pDCs. As peripheral pDCs serve a cru-
expression pattern in mTECs29. However, as these stud- cial function in the protection against viral infections
ies did not address the identity of the cross-presenting through their production of type I interferons63, the pres-
cell type, it remains to be established whether there is a ence of a pDC population in the thymus was suggested
differential contribution of resident versus migratory to reflect a similar innate immune function in a primary
cDCs in this context. lymphoid organ55.
Although DCs, on the whole, are markedly more Given the poor antigen-presenting capacity of
abundant in the medulla than in the cortex, it is unclear pDCs64, it had been considered rather unlikely that
whether this applies in equal terms to both migra- they would have a role in central tolerance; however,
tory and resident cDCs. Recent work has identified recent data suggest that this view may need to be
XC‑chemokine ligand 1 (XCL1; also known as lympho- revised. First, in a reductionist in vitro setting, thymic
tactin) as a crucial determinant of the medullary locali- pDCs were capable of presenting peptide antigen to
zation of cDCs59, as XCL1‑deficient mice have fewer specific thymocytes and promoting their differentia-
medullary cDCs. Although not directly addressed in tion into TReg cells65. Second, pDCs were reported to be
this study, the fact that only CD8α+ cDCs express the surprisingly efficient in picking up soluble or particu-
receptor for XCL1 (XCR1) suggests that this mislocaliza- late model antigens at peripheral sites and transport-
tion primarily affects resident, but not migratory, cDCs. ing them to the thymus in vivo56,66. Interestingly, CCR9,
As mTECs are the only thymic stromal cells that pro- which controls the homing of T cell progenitors to the
duce XCL1 (notably in an AIRE-dependent manner), thymus, is also crucial for pDC recruitment to the thy-
the XCL1–XCR1 chemokine axis may coordinate the mus66. A subset of CCR9+ immature pDCs in peripheral
localization of resident cDCs next to mTECs. Such a lymphoid tissues was able to promote the conversion
close apposition should facilitate the transfer of mTEC- of naive T cells into TReg cells67 and this ‘tolerogenic’
derived TRAs to DCs, although this has not yet been pDC subset may also exert tolerogenic functions upon
experimentally validated. recruitment into the thymus. Indeed, adoptively trans-
The migratory CD8α−SIRPα+ cDC subset seems ferred, ovalbumin (OVA)-loaded wild-type pDCs, but
to be guided by different cues. Thus, CC-chemokine not CCR9‑deficient pDCs, migrated to the thymus
receptor 2 (CCR2)‑deficient mice showed a selective where they specifically located to the medulla and
diminution of migratory DCs in the thymus60, whereby promoted the clonal deletion of OVA-specific OT‑II
CCR2 signalling seems to be crucial for the mobiliza- thymocytes66.
tion of peripheral SIRPα+ cDCs rather than for their Altogether, these new data present a strong case for
final intrathymic positioning. The same study showed a contribution of pDCs to central tolerance. Of note,
CCR9 that migratory cDCs can accumulate in the cortex in the in contrast to both subsets of cDCs, pDCs do not pick
CC-chemokine receptor 9; vicinity of small vessels and inside perivascular regions, up mTEC-derived antigens (J. Nedjic, T. Yamano,
a G protein-coupled receptor whereas other investigators have found that SIRPα+ J. Derbinski, L.K. and B.K., unpublished observations),
that recognizes the chemokine
CCL25 (also known as TECK
cDCs preferentially localized near blood vessels at the which indicates that they may sample self antigens in
and thymus expressed corticomedullary junction and within deeper regions the periphery and then ‘freeze’ their antigen cargo.
chemokine). of the medulla (D. Atibalentja and E. Unanue, personal Moreover, activation of Toll-like receptors (TLRs)

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 9

© 2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

prevents both cDCs and pDCs from migrating to the ‘chronic’ inappropriate help to B cells that expressed the
thymus66,68, thereby conceivably preventing central tol- respective BCR in a double-transgenic model78. This
erance to pathogens under inflammatory conditions. ultimately resulted in systemic autoimmunity 78, which
Finally, considering that CCR9 also promotes migra- indicates that robust tolerance towards this special class
tion to the intestine, CCR9+ pDCs may sample not only of self antigens is indispensable.
bona fide self antigens but also innocuous foreign anti- Cognate interactions between B cells and T cells are
gens, such as food components or constituents of the crucial for germinal centre formation during immune
commensal microflora. However, as yet there are no responses to foreign antigens, but there may be an
experimental data to support this intriguing scenario. intriguing counterpart to these interactions in the cen-
tral tolerance process. Thus, BCR-transgenic B cells effi-
Thymic B cells ciently mediated negative selection of CD4+ thymocytes
Approximately 0.3% of thymic cells are B cells, which expressing a transgenic TCR that was specific for the
is a proportion similar to that seen for DCs. The origin same cognate self antigen72. Although it may be diffi-
of thymic B cells is still a matter of debate; it is unclear cult to envisage how rare cognate self antigen-specific
whether they derive from intrathymic B lymphopoie- B cells within a polyclonal repertoire may be sufficient
sis and/or immigration of peripheral B cells69,70. The to impose tolerance, the same study reported that even
phenotypic and functional attributes of thymic B cells a polyclonal B cell population mediated a degree of
closely resemble those of conventional B cells (that is, deletion of TCR-transgenic CD4+ T cells.
B-2 cells) that are found in the periphery 71,72. However, In summary, these intriguing new data should
compared with splenic B cells, thymic B cells show reignite interest in the role of B cells in central T cell
increased expression of MHC class II and co‑stimulatory tolerance. Thymic B cells may afford another layer
molecules, which is indicative of their potent antigen- of tolerance towards BCR-derived self-constituents,
presenting capacity. Indeed, many studies have shown thus pre-empting previously described peripheral
that thymic B cells are capable of inducing negative checkpoints of T cell tolerance to this unique class of
selection. Most convincingly, myelin oligodendro- self antigens79. Other relevant issues in this regard are
cyte glycoprotein (MOG)-specific CD4+ thymocytes whether the scope of B cell-mediated central T cell
were negatively selected when an epitope of MOG was tolerance is indeed dictated by the BCR repertoire of
exclusively presented by B cells73. thymic B cells, how diverse the thymic BCR repertoire
However, it is currently unclear how thymic B cells is, and whether autoreactive B cells may be enriched in
fare in their overall contribution to tolerance when the thymus to allow for efficient presentation of soluble
compared with DCs and mTECs. For instance, dele- self antigens.
tion of superantigen-reactive CD4+ T cells was more
efficiently induced by DCs, and in the same context Perspectives
B cells entirely failed to negatively select CD8+ thy- Following our discussion of the key cell biological attrib-
mocytes74. Consistent with these in vivo data, selective utes of the different thymic APC populations in the con-
supplementation of reaggregate thymus organ cultures text of T cell repertoire selection, we close with some
(RTOCs) with different thymic APCs in the presence speculative thoughts on how the intrathymic encoun-
of soluble OVA peptide resulted in the negative selec- ter of self (or the lack thereof) may imprint peripheral
tion of OVA-specific CD4+ thymocytes when resident T cell behaviour, coordinate dominant versus reces-
or migratory cDCs were used as APCs, but not when sive mechanisms of tolerance and specify the targets of
B cells were used75. autoimmunity.
Relatively little is known about the parameters that
may shape the pMHC ligandome of thymic B cells. Positive selection, homeostatic fitness and immunity to
Traditionally, peripheral B cells are considered to be pathogens. Our discussion of antigen presentation for
poor presenters of exogenous antigens. This may also positive selection converged on the view that cTECs gen-
be the case for thymic B cells, which would explain erate and display functionally (and possibly structurally)
their poor performance in the aforementioned stud- distinct private self peptides that may sustain the selection
ies on negative selection in RTOCs. However, in con- of T cell clones that display weak tonic self-reactivity in
trast to the poor efficacy with which B cells present the periphery. This notion is at odds with the proposi-
soluble antigens, B  cell receptor (BCR)-mediated tion that the very same self peptides that mediate positive
cognate interactions lead to exceptionally efficient selection are also essential for naive T cell homeostasis
antigen presentation76. Given the copious amounts of in the periphery and act as co‑agonists when T cells
MHC class II molecules that are expressed by thymic respond to foreign antigens16,80,81.
B cells, it is conceivable that B cells may not only pre- How can this apparent discrepancy be reconciled?
sent BCR-captured external antigens but, in a man- First, it is possible that the peripheral self peptides sup-
ner similar to mTECs, they may focus their pMHC porting homeostasis and co‑activation are not identi-
class II ligandome towards endogenously expressed cal, but instead are functionally equivalent to those
proteins. This intracellular antigen pool is likely to that support positive selection. Second, one may argue
include germline-encoded or even clonotype-specific that the functional competence of the peripheral T cell
regions of the BCR77. Lack of tolerance towards vari- repertoire requires a balanced distribution of clones
able (V) regions of the BCR caused T cells to provide covering a relatively wide range of tonic self-reactivity,

10 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/immunol

© 2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

Thymus (positive selection) Periphery (steady state) Periphery (immune response)


cTEC No re-encounter of
positively selecting peptide DC Agonist
Private foreign Delayed
peptide T cell T cell peptide activation
Private
processing CD5low Less
CD5low CD5low
pathways CD3ζ AICD

MHC TCR Sustained


Strength of TCR signalling

response

CD5hi

Response
CD5low

‘Tonic’ re-encounter of
positively selecting peptide Time
cTEC DC Rapid
DC Agonist activation?
Public Public foreign
peptide peptide T cell peptide More
Public CD5hi Public AICD
CD5hi
processing processing
CD3ζ CD3ζ CD5hi
pathways pathways Less
P P sustained
response
Bystander
damage?

Figure 4 | Consequences of positive selection by ‘private’ versus ‘public’ peptides: a hypothesis. ‘Private’ peptides
(upper section of left panel) that are generated through unique proteolytic pathways in cortical Nature Reviews
thymic | Immunology
epithelial cells
(cTECs) may preferentially support the selection of CD5low T cell clones via interactions at the lower end of the affinity
range that is permissive for positive selection. One determinant of these ‘low-strength’ interactions could be that private
peptides are weak MHC binders, which is indicated here by the loose fit between the peptide and MHC (double-headed
arrows). Whether or not this is true, in the periphery, T cells that are selected on private peptides do not re‑encounter the
positively selecting peptides and hence do not receive tonic signals (middle and right panels). As a consequence, their
CD3 ζ-chains are not preloaded with basal phosphorylation. Public peptides may preferentially support the selection of
CD5hi clones via positively selecting interactions at the relatively higher end of the affinity range (lower section of left
panel). Public peptides might be good MHC binders that generate ‘low-strength’ interactions by loosely binding to the
T cell receptor (TCR; indicated by the double-headed arrow). In the periphery, continual interactions with the very same
peptides support T cell homeostasis and mediate partial CD3 ζ-chain phosphorylation (middle and right panels). During
an immune response to foreign antigens, CD5low and CD5hi T cell clones of identical specificity may differentially respond
with respect to the timing and magnitude of clonal expansion and contraction. The dominance of either type of responder
might vary with parameters such as the duration and anatomical distribution of the infection. AICD, activation-induced
cell death; DC, dendritic cell. 

as represented by CD5low and CD5hi T cells. Possibly, a determined whether these CD5hi-skewed CD8+ T cells
corresponding mix of private and public MHC ligands did indeed collapse faster, make an over-shooting
on cTECs is a prerequisite to select such a compos- pathogenic response or fail to respond to antigen at all.
ite of T cell clones with low or high tonic affinity, Against this background, it will also be interesting to
respectively. see whether the duration of infections (chronic versus
One can envisage a potential benefit of having T cells acute) or the spread of pathogens (systemic versus local)
with a wide range of affinities for self antigens (FIG. 4). are crucial determinants of the relative contribution of
Following infection with pathogens, T cells with high CD5low and CD5hi T cell clones to the immune response
affinity for self could provide a rapid, yet relatively short- to foreign antigens, and how these parameters affect their
lived, initial immune response that is then followed by a partitioning into the memory pool.
sustained response by T cells with lower self affinity. The Notwithstanding these considerations, we still lack
lower-affinity T cells are presumably not only less prone to experimental data to directly link the selection of a given
burn out but also less likely to cause bystander damage ‘low self-affinity’ TCR specificity to a specific private
to self tissues. This scenario would be consistent with the peptide, the processing of which would be dependent
observation that β5t‑deficient mice, which have a numer- on any of the cTEC-specific pathways of antigen pro-
ically smaller but presumably more strongly self-reactive cessing. Solving this issue has been hampered by our
CD8+ T cell repertoire, die in response to infection with current ignorance of the identity of the peptides bound
influenza virus11. Yet, as the influenza virus-specific to MHC that are presented by cTECs. The scarcity of
response was not tracked in this study, it remains to be cTECs (1–3 × 104 per mouse thymus) renders this a

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 11

© 2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 3 | Towards directly addressing the peptide–MHC ligandome of thymic stromal cells
Remarkably, one of the first attempts in the early 1990s to assess the peptide–MHC (pMHC) ligandome by peptide
elution and peptide sequencing reported a comparison between cortical thymic epithelial cells (cTECs) and splenic
antigen-presenting cells (APCs)113. At the time, only 17 of the most abundant self peptides had been identified, from an
estimated 2,000 to 10,000 distinct peptides presented by MHC class II or MHC class I molecules, respectively. With
technology improving, the field has seen increasingly detailed assessments of cell type-specific pMHC ligandomes,
particularly those of tumour cells. However, the ex vivo assessment of rare isolated populations, such as thymic stromal
cells, remains a major technical challenge.
In general, naturally processed peptides have been characterized from acid extractions of affinity-purified MHC
molecules, which are then sequenced, typically by mass spectrometry. This approach was used to describe peptides
bound to MHC class I and II molecules in the human thymus114,115. However, as whole tissue was used, these studies could
not assign the identified peptides to particular stromal cell types. Another recent study identified 50–100 peptides from
MHC class I or II molecules from human thymic dendritic cells (DCs) and compared these to peptides from thymic APCs
depleted of DCs116, which gave an initial insight into differences in the ligandomes of thymic APCs. The amount of
starting material required with this approach (at least 108 cells) so far precludes an informative analysis of MHC-bound
peptides from rare populations like cTECs. Another complication arises from the possibility that the elusive ‘private’
self peptides, that are presumed to be generated by the thymoproteasome, may bind more weakly to MHC class I
molecules and hence might be lost during the immunoprecipitation step. An alternative approach has been reported
that characterizes peptides that are directly extracted with mild acid elution from the surface of intact cells. As this
procedure generates a large number of peptides, not just those bound by MHC, the authors carried out a bioinformatic
comparison to peptides that were extracted from MHC-deficient cells to assign them as ‘MHC-bound’ (REF. 117).
Chemical and metabolic labelling have also been used to provide quantitative comparisons between two populations,
although this has not been applied to the thymus118.

daunting task (BOX 3). In this context, the fundamental Second, it seems unlikely that any particular modal-
issue of whether selection of a given TCR specificity ity of antigen expression — for instance, a ‘promiscuous
actually requires a single specific self peptide has not gene expression-like’ mosaic pattern versus ubiquitous
been resolved. Likewise, we do not know whether pri- expression — will invariably result in either clonal dele-
vate peptides on cTECs are equally important for CD4+ tion or clonal deviation. The view that the essential role
and CD8+ T cell repertoire selection. This question is of AIRE in mTECs can be solely ascribed to recessive
all the more interesting as high tonic self-responders tolerance through negative selection87 is difficult to rec-
among naive CD4+ T cells seem inherently more prone oncile with the ability of mTECs to autonomously induce
to undergoing peripheral conversion into induced antigen-specific TReg cells in neo-self antigen transgenic
forkhead box P3 (FOXP3)+ TReg cells82. settings34. A recent study extended the latter finding to
a naturally expressed protein by showing that an AIRE-
Modes of central tolerance: deletion versus TReg cell dif- dependent self antigen induced thymic differentiation of
ferentiation. A detailed discussion of how antigen pres- cognate TReg cells88. Intriguingly, TReg cell progenitors
entation in the thymus may operate alongside factors of unknown specificity were found to compete for the
such as TCR affinity, access to cytokines or develop- recognition of rare stimulatory TCR self ligands in the
mental tuning of thymocyte responsiveness to specify thymus89–91, which is also suggestive of a link between
clonal deletion versus clonal deviation into the FOXP3+ promiscuous gene expression in rare mTECs and TReg cell
TReg cell lineage exceeds the scope of this article (for differentiation. The possibility that the expression and
recent reviews, see REFS 83,84). Here, it may suffice to presentation of TRAs by very few mTECs may concomi-
highlight two pertinent issues. tantly induce negative selection and TReg cell induction
First, there is no evidence to indicate that any thymic has obvious implications; interactions between thymo-
APC subset is specialized to exclusively promote clonal cytes and rare APCs that present a given cognate self
deletion or TReg cell differentiation. Although efficient antigen perhaps need not be saturating after all, because
TReg cell differentiation apparently requires a properly the occasional escape of autoreactive T cells would be
formed medullary microenvironment 85, the overall size counterbalanced by dominant regulatory mechanisms.
of the polyclonal TReg cell pool is barely affected by the
specific lack of DCs or subsets of mTECs, or by aberrant Autoimmunity: what thymocytes may not see. With
MHC class II expression on these cells (reviewed in REF. 86). increasing knowledge of the multi-layered cellular and
Thus, thymic APC subsets can obviously compensate for molecular mechanisms of central tolerance, one is bound
each other as far as filling the physiological TReg cell niche to ask which parameters are most limiting and pose the
is concerned. However, considering the evidence for partly highest risk for autoimmunity. Deep sequencing data of
non-overlapping pMHC ligandomes on different thymic the mRNA signature of mTECs indicate that the scope
APCs, we deem it unlikely that this reflects a true redun- of promiscuous gene expression is even broader than
dancy at the level of TCR specificities that are recruited previously revealed by microarray analyses92. Given this
into the TReg cell repertoire. Large-scale TCR sequencing of apparently comprehensive intrathymic representation of
TReg cell repertoires in the absence of DCs or mTEC subsets otherwise tissue-specific transcriptomes, it becomes even
is expected to resolve this issue. more intriguing and informative to know which fraction

12 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/immunol

© 2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

of self is actually not covered by central tolerance. Any expression and may thus be a risk factor for autoim-
advances in understanding the molecular regulation munity54. Finally, post-translational modifications of
of promiscuous gene expression by AIRE and beyond autoantigens that affect T cell epitopes by cell-type
would help us to understand how the tolerizing pMHC specific expression of modifying enzymes in periph-
ligandome in the thymus is generated in the first place eral target tissues, but not mTECs, may have the same
and, importantly, how dysregulation at the genetic, epi- effect. Examples of this include glycosylation of colla-
genetic and biochemical level could undermine central gen type II and citrullination of aggrecan and vimentin
tolerance and thus possibly explain immune pathologies. in the case of rheumatoid arthritis104, or deamidation of
In fact, an increasing number of examples show how insulin in the case of type 1 diabetes105.
‘holes’ in central tolerance might specify dominant T cell
targets in organ-specific autoimmune diseases. Conclusions
First, very low expression levels of particular The affinity model remains a useful framework to con-
self antigens in human mTECs correlate with a high ceptualize thymic selection events and there has been
frequency of cognate autoreactive T cells and auto­ considerable progress in understanding how the TCR
antibodies in the case of the myosin heavy chain 6 relays quantitative differences in signal strength into
(REFS 93,94), which is an autoantigen in autoimmune qualitatively different cell fates (reviewed in REF. 106).
myocarditis, or glutamate decarboxylase 2 (GAD2; Yet, as reviewed here, a coherent model of thymocyte
also known as GAD65) (REF. 95) and zinc transporter 8 selection will also need to consider temporal, spatial and
(ZNT8), which are both targets in type 1 diabetes (B.K., qualitative aspects of self recognition in distinct thymic
S. Pinto and R. Mallone, unpublished observations). microenvironments. In terms of positive selection, it
Genetic polymorphisms in the regulatory regions of remains tantalizing why ‘monogamous’ interactions
some autoantigens — which exert subtle effects on between developing T cells and a single ‘dedicated’ stro-
the expression level of self antigens in mTECs but not mal cell type are so important, and we have discussed
in peripheral tissues — correlate with susceptibility how this crucial role of cTECs may be specified by their
to organ-specific autoimmunity, as shown for insu- ability to generate ‘private’ pMHC ligands. With regard to
lin and type 1 diabetes96–98, the α‑chain of the acetyl- central tolerance induction, it is intuitively obvious why
choline receptor and myasthenia gravis99, and for the ‘promiscuity’ — not only at the level of TRA expression
thyroid-stimulating hormone receptor and Graves’ in mTECs but also in terms of the diverse characteris-
disease100. Second, similar effects are observed when tics and sheer number of contributing APC subsets — is
dominant T cell epitopes are only present in peripheral advantageous. Unravelling functional non-redundancies
tissues but not in the thymus either owing to differen- between tolerogenic APCs, at the level of the self anti-
tial splicing 101,102 or owing to ‘mis-initiation’ of mRNA gen spectra that are presented and regarding tolerance
transcription in mTECs103. Mis-initiated transcription mechanisms (clonal deletion versus TReg cell induction),
is possibly a more general feature of promiscuous gene remains a major challenge for future investigations.

1. Kyewski, B. & Klein, L. A central role for central 11. Nitta, T. et al. Thymoproteasome shapes interaction. Reference 18 concludes that T cells with
tolerance. Annu. Rev. Immunol. 24, 571–606 immunocompetent repertoire of CD8+ T cells. stronger affinity for self dominate in response to
(2006). Immunity 32, 29–40 (2010). infections, whereas reference 19 challenges the
2. Nakagawa, Y. et al. Thymic nurse cells provide 12. Xing, Y., Jameson, S. C. & Hogquist, K. A. generality of such correlations.
microenvironment for secondary T cell receptor-α Thymoproteasome subunit‑β5T generates peptide- 20. Persaud, S. P., Parker, C. R., Lo, W. L., Weber, K. S. &
rearrangement in cortical thymocytes. Proc. Natl MHC complexes specialized for positive selection. Allen, P. M. Intrinsic CD4+ T cell sensitivity and
Acad. Sci. USA 109, 20572–20577 (2012). Proc. Natl Acad. Sci. USA 110, 6979–6984 (2013). response to a pathogen are set and sustained by avidity
3. Klein, L., Hinterberger, M., Wirnsberger, G. & 13. Ziegler, A., Muller, C. A., Bockmann, R. A. & for thymic and peripheral complexes of self peptide and
Kyewski, B. Antigen presentation in the thymus for Uchanska-Ziegler, B. Low-affinity peptides and T‑cell MHC. Nature Immunol. 15, 266–274 (2014).
positive selection and central tolerance induction. selection. Trends Immunol. 30, 53–60 (2009). 21. Surh, C. D. & Sprent, J. T‑cell apoptosis detected
Nature Rev. Immunol. 9, 833–844 (2009). 14. Ryan, K. R., McNeil, L. K., Dao, C., Jensen, P. E. & in situ during positive and negative selection in the
4. Florea, B. I. et al. Activity-based profiling reveals Evavold, B. D. Modification of peptide interaction with thymus. Nature 372, 100–103 (1994).
reactivity of the murine thymoproteasome-specific MHC creates TCR partial agonists. Cell. Immunol. 22. Daley, S. R., Hu, D. Y. & Goodnow, C. C. Helios marks
subunit β5t. Chem. Biol. 17, 795–801 (2010). 227, 70–78 (2004). strongly autoreactive CD4+ T cells in two major waves
5. Murata, S. et al. Regulation of CD8+ T cell 15. Azzam, H. S. et al. CD5 expression is developmentally of thymic deletion distinguished by induction of PD‑1
development by thymus-specific proteasomes. regulated by T cell receptor (TCR) signals and TCR or NF‑κB. J. Exp. Med. 210, 269–285 (2013).
Science 316, 1349–1353 (2007). avidity. J. Exp. Med. 188, 2301–2311 (1998). 23. Stritesky, G. L. et al. Murine thymic selection quantified
6. Nakagawa, T. et al. Cathepsin L: critical role in Ii 16. Stefanova, I., Dorfman, J. R. & Germain, R. N. using a unique method to capture deleted T cells.
degradation and CD4 T cell selection in the thymus. Self-recognition promotes the foreign antigen sensitivity Proc. Natl Acad. Sci. USA 110, 4679–4684 (2013).
Science 280, 450–453 (1998). of naive T lymphocytes. Nature 420, 429–434 (2002). Using different approaches, references 22 and 23
7. Gommeaux, J. et al. Thymus-specific serine protease 17. Cho, J. H., Kim, H. O., Surh, C. D. & Sprent, J. quantify ‘early’ and ‘late’ negative selection in the
regulates positive selection of a subset of CD4+ T cell receptor-dependent regulation of lipid rafts cortex and the medulla, respectively, and conclude
thymocytes. Eur. J. Immunol. 39, 956–964 (2009). controls naive CD8+ T cell homeostasis. Immunity that the extent of clonal deletion in the cortex
8. Nedjic, J., Aichinger, M., Mizushima, N. & Klein, L. 32, 214–226 (2010). exceeds that in the medulla.
Macroautophagy, endogenous MHC II loading and 18. Palmer, M. J., Mahajan, V. S., Chen, J., Irvine, D. J. & 24. McCaughtry, T. M., Baldwin, T. A., Wilken, M. S. &
T cell selection: the benefits of breaking the rules. Lauffenburger, D. A. Signaling thresholds govern Hogquist, K. A. Clonal deletion of thymocytes can
Curr. Opin. Immunol. 21, 92–97 (2009). heterogeneity in IL‑7‑receptor-mediated responses of occur in the cortex with no involvement of the
9. Nedjic, J., Aichinger, M., Emmerich, J., Mizushima, N. naive CD8+ T cells. Immunol. Cell Biol. 89, 581–594 medulla. J. Exp. Med. 205, 2575–2584 (2008).
& Klein, L. Autophagy in thymic epithelium shapes (2011). 25. Melichar, H. J., Ross, J. O., Herzmark, P.,
the T‑cell repertoire and is essential for tolerance. 19. Mandl, J. N., Monteiro, J. P., Vrisekoop, N. & Hogquist, K. A. & Robey, E. A. Distinct temporal
Nature 455, 396–400 (2008). Germain, R. N. T cell-positive selection uses self-ligand patterns of T cell receptor signaling during positive
10. Honey, K., Nakagawa, T., Peters, C. & Rudensky, A. binding strength to optimize repertoire recognition of versus negative selection in situ. Sci. Signal. 6, ra92
Cathepsin L regulates CD4+ T cell selection foreign antigens. Immunity 38, 263–274 (2013). (2013).
independently of its effect on invariant chain: a role in References 18 and 19 show that T cell responsiveness 26. Irla, M., Hollander, G. & Reith, W. Control of central
the generation of positively selecting peptide ligands. is set in the thymus and maintained in mature T cells self-tolerance induction by autoreactive CD4+
J. Exp. Med. 195, 1349–1358 (2002). in proportion to the avidity of the positively selecting thymocytes. Trends Immunol. 31, 71–79 (2010).

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 13

© 2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

27. Mathis, D. & Benoist, C. Aire. Annu. Rev. Immunol. 51. Klein, L. Dead man walking: how thymocytes scan the negative selection. Proc. Natl Acad. Sci. USA 110,
27, 287–312 (2009). medulla. Nature Immunol. 10, 809–811 (2009). 17011–17016 (2013).
28. Peterson, P., Org, T. & Rebane, A. Transcriptional 52. Derbinski, J., Pinto, S., Rosch, S., Hexel, K. & Using BCR- and TCR-transgenic mice, this study
regulation by AIRE: molecular mechanisms of central Kyewski, B. Promiscuous gene expression patterns in shows that autoreactive thymic B cells are efficient
tolerance. Nature Rev. Immunol. 8, 948–957 (2008). single medullary thymic epithelial cells argue for a APCs for negative selection. Thymic B cells may
29. Gallegos, A. M. & Bevan, M. J. Central tolerance to stochastic mechanism. Proc. Natl Acad. Sci. USA 105, capture autoantigens through their BCR and present
tissue-specific antigens mediated by direct and 657–662 (2008). these to developing thymocytes for clonal deletion.
indirect antigen presentation. J. Exp. Med. 200, 53. Pinto, S. et al. Overlapping gene coexpression 73. Frommer, F. & Waisman, A. B cells participate in
1039–1049 (2004). patterns in human medullary thymic epithelial cells thymic negative selection of murine auto-reactive
30. Oukka, M., Cohen-Tannoudji, M., Tanaka, Y., generate self-antigen diversity. Proc. Natl Acad. Sci. CD4+ T cells. PLoS ONE 5, e15372 (2010).
Babinet, C. & Kosmatopoulos, K. Medullary thymic USA 110, E3497–3505 (2013). 74. Kleindienst, P., Chretien, I., Winkler, T. & Brocker, T.
epithelial cells induce tolerance to intracellular 54. Villasenor, J., Besse, W., Benoist, C. & Mathis, D. Functional comparison of thymic B cells and dendritic
proteins. J. Immunol. 156, 968–975 (1996). Ectopic expression of peripheral-tissue antigens in the cells in vivo. Blood 95, 2610–2616 (2000).
31. Hinterberger, M. et al. Autonomous role of medullary thymic epithelium: probabilistic, monoallelic, 75. Guerri, L. et al. Analysis of APC types involved in
thymic epithelial cells in central CD4+ T cell tolerance. misinitiated. Proc. Natl Acad. Sci. USA 105, CD4 tolerance and regulatory T cell generation using
Nature Immunol. 11, 512–519 (2010). 15854–15859 (2008). reaggregated thymic organ cultures. J. Immunol. 190,
Through diminution of MHC class II on mTECs, 55. Wu, L. & Shortman, K. Heterogeneity of thymic 2102–2110 (2013).
this study documents an autonomous contribution dendritic cells. Semin. Immunol. 17, 304–312 76. Yuseff, M. I., Pierobon, P., Reversat, A. &
of mTECs to both dominant and recessive (2005). Lennon-Dumenil, A. M. How B cells capture, process
mechanisms of CD4+ T cell tolerance and provides 56. Li, J., Park, J., Foss, D. & Goldschneider, I. and present antigens: a crucial role for cell polarity.
experimental support for the affinity model of Thymus‑homing peripheral dendritic cells constitute Nature Rev. Immunol. 13, 475–486 (2013).
TReg cell development versus clonal deletion. two of the three major subsets of dendritic cells in the 77. Weiss, S. & Bogen, B. MHC class II‑restricted
32. Klein, L., Klein, T., Ruther, U. & Kyewski, B. CD4 T cell steady-state thymus. J. Exp. Med. 206, 607–622 presentation of intracellular antigen. Cell 64,
tolerance to human C‑reactive protein, an inducible (2009). 767–776 (1991).
serum protein, is mediated by medullary thymic 57. Joffre, O. P., Segura, E., Savina, A. & Amigorena, S. 78. Munthe, L. A., Corthay, A., Os, A., Zangani, M. &
epithelium. J. Exp. Med. 188, 5–16 (1998). Cross-presentation by dendritic cells. Nature Rev. Bogen, B. Systemic autoimmune disease caused by
33. Oukka, M. et al. CD4 T cell tolerance to nuclear Immunol. 12, 557–569 (2012). autoreactive B cells that receive chronic help from Ig V
proteins induced by medullary thymic epithelium. 58. Proietto, A. I., Lahoud, M. H. & Wu, L. Distinct region-specific T cells. J. Immunol. 175, 2391–2400
Immunity 4, 545–553 (1996). functional capacities of mouse thymic and splenic (2005).
34. Aschenbrenner, K. et al. Selection of Foxp3+ dendritic cell populations Immunol. Cell Biol. 86, 79. Detanico, T., Heiser, R. A., Aviszus, K., Bonorino, C. &
regulatory T cells specific for self antigen expressed 700–708 (2008). Wysocki, L. J. Self-tolerance checkpoints in CD4 T cells
and presented by Aire+ medullary thymic epithelial 59. Lei, Y. et al. Aire-dependent production of XCL1 specific for a peptide derived from the B cell antigen
cells. Nature Immunol. 8, 351–358 (2007). mediates medullary accumulation of thymic dendritic receptor. J. Immunol. 187, 82–91 (2011).
35. Atibalentja, D. F., Byersdorfer, C. A. & Unanue, E. R. cells and contributes to regulatory T cell development. 80. Ebert, P. J., Jiang, S., Xie, J., Li, Q. J. & Davis, M. M.
Thymus-blood protein interactions are highly effective J. Exp. Med. 208, 383–394 (2011). An endogenous positively selecting peptide
in negative selection and regulatory T cell induction. 60. Baba, T., Nakamoto, Y. & Mukaida, N. Crucial enhances mature T cell responses and becomes an
J. Immunol. 183, 7909–7918 (2009). contribution of thymic Sirpα+ conventional dendritic autoantigen in the absence of microRNA miR‑181a.
36. Klein, L., Roettinger, B. & Kyewski, B. Sampling of cells to central tolerance against blood-borne antigens Nature Immunol. 10, 1162–1169 (2009).
complementing self-antigen pools by thymic stromal in a CCR2‑dependent manner. J. Immunol. 183, 81. Lo, W. L. et al. An endogenous peptide positively
cells maximizes the scope of central T cell tolerance. 3053–3063 (2009). selects and augments the activation and survival of
Eur. J. Immunol. 31, 2476–2486 (2001). 61. Atibalentja, D. F., Murphy, K. M. & Unanue, E. R. peripheral CD4+ T cells. Nature Immunol. 10,
37. Munz, C. Enhancing immunity through autophagy. Functional redundancy between thymic CD8α+ and 1155–1161 (2009).
Annu. Rev. Immunol. 27, 423–449 (2009). Sirpα+ conventional dendritic cells in presentation of 82. Martin, B. et al. Highly self-reactive naive CD4 T cells
38. Aichinger, M., Wu, C., Nedjic, J. & Klein, L. blood-derived lysozyme by MHC class II proteins. are prone to differentiate into regulatory T cells.
Macroautophagy substrates are loaded onto J. Immunol. 186, 1421–1431 (2011). Nature Commun. 4, 2209 (2013).
MHC class II of medullary thymic epithelial cells for 62. Baba, T., Badr Mel, S., Tomaru, U., Ishizu, A. & 83. Hsieh, C. S., Lee, H. M. & Lio, C. W. Selection of
central tolerance. J. Exp. Med. 210, 287–300 Mukaida, N. Novel process of intrathymic tumor- regulatory T cells in the thymus. Nature Rev. Immunol.
(2013). immune tolerance through CCR2‑mediated 12, 157–167 (2012).
39. Mizushima, N. Autophagy in protein and organelle recruitment of Sirpα+ dendritic cells: a murine model. 84. Wirnsberger, G., Hinterberger, M. & Klein, L.
turnover. Cold Spring Harb. Symp. Quant. Biol. 76, PLoS ONE 7, e41154 (2012). Regulatory T‑cell differentiation versus clonal deletion
397–402 (2011). 63. Reizis, B., Colonna, M., Trinchieri, G., Barrat, F. & of autoreactive thymocytes. Immunol. Cell Biol. 89,
40. Dongre, A. R. et al. In vivo MHC class II presentation Gilliet, M. Plasmacytoid dendritic cells: one-trick 45–53 (2011).
of cytosolic proteins revealed by rapid automated ponies or workhorses of the immune system? 85. Cowan, J. E. et al. The thymic medulla is required
tandem mass spectrometry and functional analyses. Nature Rev. Immunol. 11, 558–565 (2011). for Foxp3+ regulatory but not conventional CD4+
Eur. J. Immunol. 31, 1485–1494 (2001). 64. Villadangos, J. A. & Young, L. Antigen-presentation thymocyte development. J. Exp. Med. 210, 675–681
41. Klein, L., Hinterberger, M., von Rohrscheidt, J. & properties of plasmacytoid dendritic cells. Immunity (2013).
Aichinger, M. Autonomous versus dendritic cell- 29, 352–361 (2008). 86. Klein, L. & Jovanovic, K. Regulatory T cell lineage
dependent contributions of medullary thymic 65. Wirnsberger, G., Mair, F. & Klein, L. Regulatory T cell commitment in the thymus. Semin. Immunol. 23,
epithelial cells to central tolerance. Trends Immunol. differentiation of thymocytes does not require a 401–409 (2011).
32, 188–193 (2011). dedicated antigen-presenting cell but is under T cell- 87. Mathis, D. & Benoist, C. A decade of AIRE.
42. Koble, C. & Kyewski, B. The thymic medulla: a unique intrinsic developmental control. Proc. Natl Acad. Sci. Nature Rev. Immunol. 7, 645–650 (2007).
microenvironment for intercellular self-antigen USA 106, 10278–10283 (2009). 88. Malchow, S. et al. Aire-dependent thymic development
transfer. J. Exp. Med. 206, 1505–1513 (2009). 66. Hadeiba, H. et al. Plasmacytoid dendritic cells of tumor-associated regulatory T cells. Science 339,
43. Hubert, F. X. et al. Aire regulates the transfer of transport peripheral antigens to the thymus to 1219–1224 (2013).
antigen from mTECs to dendritic cells for induction of promote central tolerance. Immunity 36, 438–450 This study reports that TReg cells that were
thymic tolerance. Blood 118, 2462–2472 (2011). (2012). consistently found to be enriched in prostate
44. Taniguchi, R. T. et al. Detection of an autoreactive This study shows that endogenous pDCs take up tumours of mice recognized an unknown antigen
T‑cell population within the polyclonal repertoire that subcutaneously injected antigen and transport that was also present in the healthy prostate.
undergoes distinct autoimmune regulator (Aire)- it to the thymus in a CCR9‑dependent manner. These cells were found to differentiate as ‘natural’
mediated selection. Proc. Natl Acad. Sci. USA 109, Upon intravenous injection, antigen-loaded pDCs (that is, thymically induced) TReg cells in an
7847–7852 (2012). delete specific thymocytes, which indicates that AIRE-dependent manner, which provides evidence
45. Irla, M. et al. Autoantigen-specific interactions with migratory pDCs can support central tolerance. for a link between AIRE-mediated expression of
CD4+ thymocytes control mature medullary thymic 67. Hadeiba, H. et al. CCR9 expression defines peripheral tissue antigens and the development of
epithelial cell cellularity. Immunity 29, 451–463 tolerogenic plasmacytoid dendritic cells able to organ-specific TReg cells.
(2008). suppress acute graft-versus-host disease. Nature 89. Bautista, J. L. et al. Intraclonal competition limits the
46. DeVoss, J. et al. Spontaneous autoimmunity Immunol. 9, 1253–1260 (2008). fate determination of regulatory T cells in the thymus.
prevented by thymic expression of a single self- 68. Bonasio, R. et al. Clonal deletion of thymocytes by Nature Immunol. 10, 610–617 (2009).
antigen. J. Exp. Med. 203, 2727–2735 (2006). circulating dendritic cells homing to the thymus. 90. Leung, M. W., Shen, S. & Lafaille, J. J. TCR-dependent
47. Fan, Y. et al. Thymus-specific deletion of insulin Nature Immunol. 7, 1092–1100 (2006). differentiation of thymic Foxp3+ cells is limited to
induces autoimmune diabetes. EMBO J. 28, 69. Akashi, K., Richie, L. I., Miyamoto, T., Carr, W. H. & small clonal sizes. J. Exp. Med. 206, 2121–2130
2812–2824 (2009). Weissman, I. L. B lymphopoiesis in the thymus. (2009).
48. Ehrlich, L. I., Oh, D. Y., Weissman, I. L. & Lewis, R. S. J. Immunol. 164, 5221–5226 (2000). 91. Moran, A. E. et al. T cell receptor signal strength in
Differential contribution of chemotaxis and substrate 70. Feyerabend, T. B. et al. Deletion of Notch1 converts Treg and iNKT cell development demonstrated by a
restriction to segregation of immature and mature pro‑T cells to dendritic cells and promotes thymic novel fluorescent reporter mouse. J. Exp. Med. 208,
thymocytes. Immunity 31, 986–998 (2009). B cells by cell-extrinsic and cell-intrinsic mechanisms. 1279–1289 (2011).
49. Le Borgne, M. et al. The impact of negative selection Immunity 30, 67–79 (2009). 92. St‑Pierre, C. et al. Transcriptome sequencing of
on thymocyte migration in the medulla. Nature 71. Mori, S. et al. Presence of B cell progenitors in the neonatal thymic epithelial cells. Sci. Rep. 3, 1860
Immunol. 10, 823–830 (2009). thymus. J. Immunol. 158, 4193–4199 (1997). (2013).
50. Ueda, Y. et al. Mst1 regulates integrin-dependent 72. Perera, J., Meng, L., Meng, F. & Huang, H. 93. Lv, H. et al. Impaired thymic tolerance to α-myosin
thymocyte trafficking and antigen recognition in the Autoreactive thymic B cells are efficient antigen- directs autoimmunity to the heart in mice and
thymus. Nature Commun. 3, 1098 (2012). presenting cells of cognate self-antigens for T cell humans. J. Clin. Invest. 121, 1561–1573 (2011).

14 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/immunol

© 2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

94. Gottumukkala, R. V. et al. Myocardial infarction 103. Pinto, S. et al. Mis-initiation of intrathymic MART-1 113. Marrack, P., Ignatowicz, L., Kappler, J. W., Boymel, J.
triggers chronic cardiac autoimmunity in type 1 transcription and biased TCR usage explain the high & Freed, J. H. Comparison of peptides bound to spleen
diabetes. Sci. Transl Med. 4, 138ra180 (2012). frequency of MART-1-specific T cells. Eur. J. Immunol. and thymus class II. J. Exp. Med. 178, 2173–2183
95. Gotter, J., Brors, B., Hergenhahn, M. & Kyewski, B. (in the press). (1993).
Medullary epithelial cells of the human thymus 104. Scally, S. W. et al. A molecular basis for the 114. Collado, J. A. et al. Composition of the
express a highly diverse selection of tissue-specific association of the HLA‑DRB1 locus, citrullination, HLA‑DR‑associated human thymus peptidome.
genes colocalized in chromosomal clusters. and rheumatoid arthritis. J. Exp. Med. 210, Eur. J. Immunol. 43, 2273–2282 (2013).
J. Exp. Med. 199, 155–166 (2004). 2569–2582 (2013). 115. Espinosa, G. et al. Peptides presented by HLA class I
96. Durinovic-Bello, I. et al. Insulin gene VNTR genotype 105. van Lummel, M. et al. Post-translational modification molecules in the human thymus. J. Proteomics 94,
associates with frequency and phenotype of the of HLA-DQ binding islet-autoantigens in type 1 23–36 (2013).
autoimmune response to proinsulin. Genes Immun. diabetes. Diabetes 63, 237–247 (2014). 116. Adamopoulou, E. et al. Exploring the MHC-peptide
11, 188–193 (2010). 106. Gascoigne, N. R. & Palmer, E. Signaling in thymic matrix of central tolerance in the human thymus.
97. Pugliese, A. et al. The insulin gene is transcribed in selection. Curr. Opin. Immunol. 23, 207–212 (2011). Nature Commun. 4, 2039 (2013).
the human thymus and transcription levels correlated 107. Bains, I., van Santen, H. M., Seddon, B. & Yates, A. J. 117. Fortier, M. H. et al. The MHC class I peptide repertoire
with allelic variation at the INS VNTR‑IDDM2 Models of self-peptide sampling by developing T cells is molded by the transcriptome. J. Exp. Med. 205,
susceptibility locus for type 1 diabetes. Nature Genet. identify candidate mechanisms of thymic selection. 595–610 (2008).
15, 293–297 (1997). PLoS Comput. Biol. 9, e1003102 (2013). 118. Mester, G., Hoffmann, V. & Stevanovic, S. Insights into
98. Vafiadis, P. et al. Insulin expression in human thymus 108. Org, T. et al. The autoimmune regulator PHD finger MHC class I antigen processing gained from large-
is modulated by INS VNTR alleles at the IDDM2 locus. binds to non-methylated histone H3K4 to activate scale analysis of class I ligands. Cell. Mol. Life Sci. 68,
Nature Genet. 15, 289–292 (1997). gene expression. EMBO Rep. 9, 370–376 (2008). 1521–1532 (2011).
99. Giraud, M. et al. An IRF8‑binding promoter variant 109. Koh, A. S. et al. Aire employs a histone-binding 119. Millet, V., Naquet, P. & Guinamard, R. R. Intercellular
and AIRE control CHRNA1 promiscuous expression in module to mediate immunological tolerance, linking MHC transfer between thymic epithelial and dendritic
thymus. Nature 448, 934–937 (2007). chromatin regulation with organ-specific cells. Eur. J. Immunol. 38, 1257–1263 (2008).
100. Colobran, R. et al. Association of an SNP with autoimmunity. Proc. Natl Acad. Sci. USA 105,
intrathymic transcription of TSHR and Graves’ disease: 15878–15883 (2008). Acknowledgements
a role for defective thymic tolerance. Hum. Mol. Genet. 110. Abramson, J., Giraud, M., Benoist, C. & Mathis, D. L.K. received support from the Deutsche
20, 3415–3423 (2011). Aire’s partners in the molecular control of Forschungsgemeinschaft, Germany (Collaborative research
101. Klein, L., Klugmann, M., Nave, K. A., Tuohy, V. K. & immunological tolerance. Cell 140, 123–135 centre SFB 1054 and grants KL 1228/4‑1 and KL 1228/5‑1).
Kyewski, B. Shaping of the autoreactive T‑cell (2010). B.K. was supported by the German Cancer Research Center
repertoire by a splice variant of self protein expressed 111. Giraud, M. et al. Aire unleashes stalled RNA (DKFZ), the Deutsche Forschungsgemeinschaft (Collaborative
in thymic epithelial cells. Nature Med. 6, 56–61 (2000). polymerase to induce ectopic gene expression in research centre SFB 938) and the European Research
102. de Jong, V. M. et al. Alternative splicing and thymic epithelial cells. Proc. Natl Acad. Sci. USA 109, Council (ERC‑2012‑AdG). P.M.A. was supported by the US
differential expression of the islet autoantigen IGRP 535–540 (2012). National Institutes of Health (NIH) grant AI‑24157. K.A.H is
between pancreas and thymus contributes to 112. Danso-Abeam, D., Humblet-Baron, S., Dooley, J. & supported by NIH grants AI088209, AI35296 and AI39560.
immunogenicity of pancreatic islets but not Liston, A. Models of aire-dependent gene regulation
diabetogenicity in humans. Diabetologia 56, for thymic negative selection. Frontiers Immunol. 2, Competing interests statement
2651–2658 (2013). 14 (2011). The authors declare no competing interests.

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 15

© 2014 Macmillan Publishers Limited. All rights reserved

You might also like