You are on page 1of 5

A RTICLES

Aire regulates negative selection of


organ-specific T cells
Adrian Liston1, Sylvie Lesage1, Judith Wilson1, Leena Peltonen2 and Christopher C. Goodnow1
© 2003 Nature Publishing Group http://www.nature.com/natureimmunology

Published online 3 March 2003; doi:10.1038/ni906

Autoimmune polyendocrinopathy syndrome type 1 is a recessive Mendelian disorder resulting from


mutations in a novel gene, AIRE, and is characterized by a spectrum of organ-specific autoimmune
diseases. It is not known what tolerance mechanisms are defective as a result of AIRE mutation. By
tracing the fate of autoreactive CD4 + T cells with high affinity for a pancreatic antigen in transgenic
mice with an Aire mutation, we show here that Aire deficiency causes almost complete failure to
delete the organ-specific cells in the thymus.These results indicate that autoimmune polyendocrino-
pathy syndrome 1 is caused by failure of a specialized mechanism for deleting forbidden T cell clones,
establishing a central role for this tolerance mechanism.

Immunological tolerance to organ-specific antigens poses a particular induce tolerance. Organ-specific T cells in several different lines of T
problem. These antigens are often present in high concentrations in cell receptor (TCR) transgenic mice have a complete lack of central
peripheral organs, where they potentially activate both high- and low- or peripheral tolerance1–6. For these reasons, it is thought that organ-
affinity T cells, but only trace amounts occur in the circulation, specific tolerance depends primarily on a passive process of immuno-
peripheral lymphoid tissues or thymus, where they are needed to logical ‘ignorance’ due to sequestration of the antigen from naive T
cells1, or on peripheral tolerance mechanisms such as peripheral dele-
tion7–9, anergy9, suppression by regulatory T cells10,11 or other, less
a b c d well-defined, regulatory mechanisms4,5. A growing body of findings,
however, indicates that the thymus may have an impact on organ-spe-
cific tolerance. When high-affinity T cells recognizing dominant pep-
tide epitopes of model antigens were tracked in TCR transgenic mice
expressing these antigens under organ-specific promoters, the pre-
vailing outcomes are thymic clonal deletion and anergy of the autore-
active T cells12–15. Analysis of normal and transgenic mice showed that
a wide range of ‘organ-specific antigens’ are actually expressed by
rare peripheral antigen expressing (PAE) thymic medullary epithelial
cells16,17. Thus, there are widely divergent views about normal resis-
tance to organ-specific autoimmune diseases.
Although transgenic animal models have been crucial in establish-
ing the possible fates of autoreactive T cells, resolving the divergent
mechanisms of tolerance requires ways to test their significance for
immune regulation in humans and animals with normal lymphocyte
repertoires. Analysis of genetic defects that result in spontaneous
autoimmune disease in humans provides, in principal, a powerful
bridge between human immunoregulation and the transgenic mouse

Figure 1. Failure to delete pancreatic islet reactive T cells in thymus of


Aire–/– mice. T cells bearing the insHEL-reactive TCR were stained with 1G12 anti-
TCR clonotypic antibody and T cell maturation markers and analyzed by four-color
flow cytometry. Representative profiles from mice of the indicated genotypes are
shown. Numbers indicate the percentages of gated cells that fall within the indicated
regions. Histogram profiles show the control B10.Br Aire+/+ TCR transgenic mice (in
white) superimposed on the indicated profile (in gray) to facilitate comparison. (a)
Control B10.Br Aire+/+ TCR transgenic mice, lacking the insHEL gene. (b) Control
B10.Br Aire+/+ TCR-insHEL double-transgenic mice. (c) Aire+/– TCR-insHEL double-
transgenic mice. (d) Aire–/– TCR-insHEL double-transgenic mice.

1
ACRF Genetics Lab, Medical Genome Centre, John Curtin School of Medical Research,The Australian National University, ACT 2601 Australia. 2Department of Medical
Genetics and Molecular Medicine, University of Helsinki and National Public Health Institute, Finland. Correspondence should be addressed to C.C.G.
(chris.goodnow@anu.edu.au).

350 nature immunology • volume 4 no 4 • april 2003 • www.nature.com/natureimmunology


A RTICLES

Table 1. Loss of Aire rescues autoreactive CD4 T cells from negative selection
Aire genotype B10.Br B10.Br Aire+/+ Aire+/– Aire–/–
Transgene genotype TCR+ TCR+HEL+ TCR+HEL+ TCR+HEL+ TCR+HEL+

CD4+CD8+double positive cellsa 680 ± 130 270 ± 100 270 ± 140 500 ± 230 680 ± 130
CD4+CD8– single positive cellsa 360 ± 60 50 ± 20 44 ± 20 98 ± 40 230 ± 80
CD4+CD8– 1G12hi cellsa † 220 ± 86 9.0 ± 3 4.8 ± 4 39 ± 21 120 ± 42
© 2003 Nature Publishing Group http://www.nature.com/natureimmunology

CD4+CD8– 1G12hi CD69– cellsa 110 ± 51 2.8 ± 1 1.3 ± 1 15 ± 10 61 ± 34


CD4+CD8– 1G12hi CCL19-Rhi cellsa 110 ± 43 1.9 ± 2 0.9 ± 0.8 11 ± 10 59 ± 34
CD4+CD8– 1G12+CD25hi cellsa 7.7 ± 4 8.2 ± 3 4.1 ± 3 7.3 ± 2 3.8 ± 0.7
CD25hi within CD4+CD8– 1G12+ cells (%) 2.3 ± 0.8 24.5 ± 3 16 ± 4 9.7 ± 0.4 2.2 ± 0.7
Mean CD5 on CD4+CD8– 1G12+ cells (relative to TCR+) 100 152 ± 33 129 ± 5 133 ± 12 109 ± 9
Mean CD3 on CD4+CD8– 1G12+ cells (relative to TCR+) 100 50 ± 12 51 ± 9 70 ± 15 86 ± 13
1G12+CD4+ splenocytes (%) 6.1 ± 2.2 0.4 ± 0.1 0.4 ± 0.1 0.6 ± 0.3 2.2 ± 2

Mean ± s.d. aCell no. × 105.

models needed to analyze tolerance mechanisms. A key opportunity is avidity and specificity for a dominant peptide of hen egg lysozyme (HEL)
the genetic defect responsible for human autoimmune polyen- complexed with the major histocompatibility molecule (MHC) I-Ak (ref.
docrinopathy syndrome type 1, a rare recessive mendelian disorder 28). HEL induces immunological tolerance when expressed as a neo-self
characterized by a spectrum of autoimmune diseases involving multi- antigen from transgenes controlled by systemic or organ-specific promot-
ple peripheral organs (reviewed at http://www3.ncbi.nlm.nih.gov, ers29,30. When the membrane HEL transgene is controlled by the rat insulin
OMIM 240300). Common conditions include various autoimmune promoter (insHEL), high concentrations of antigen are expressed in the β
endocrinopathies, gastrointestinal disorders, skin diseases and chronic cells of the pancreatic islet and low concentrations (comparable to those
hepatitis, with autoantibodies produced to many organ-specific of insulin) circulate systemically14. In TCR-insHEL double-transgenic
autoantigens18. The disease results from loss-of-function mutations in mice, 3A9 T cells are efficiently tolerized despite their high frequency,
the gene AIRE19,20. The AIRE protein seems to act as a novel tran- through deletion of high-avidity T cells in the thymus and export of resid-
scriptional regulator21,22 and is chiefly expressed in rare thymus stro- ual cells that are functionally anergic and express lower amounts of HEL-
mal cells23–25. An apt model of human autoimmune polyendocrinopa- specific TCR and CD3 and increased amounts of CD514,27. We show here
thy syndrome is the Aire-mutant mouse, which shows a range of that Aire mutation causes almost complete failure to delete pancreatic
autoimmune disorders similar to that observed in humans23. Analysis islet–reactive T cells in the thymus, directly linking autoimmune polyen-
of Aire-deficient mice showed that Aire is required for low-level docrinopathy syndrome 1 to failure of thymic deletion and establishing a
thymic epithelial expression of a subset of organ-specific mRNAs, central role for this mechanism in immune regulation.
although only a minority of the mRNAs expressed ectopically in
thymic medullary epithelial cells seemed to be dependent on Aire26. Results
Resolving the role of Aire in organ-specific tolerance awaits a deter- Aire and central tolerance
mination of what, if any, T cell tolerance mechanisms are disrupted by To determine whether or not Aire has a role in T cell tolerance mecha-
the gene defect, particularly because thymic medullary expression of nisms, Aire–/– mice were crossed to 3A9 TCR-insHEL transgenic ani-
one of the best-characterized examples of a peripheral antigen that mals, enabling direct and sensitive tracking of islet-specific T cells.
triggers thymic deletion, the liver C-reactive protein15, is not depen- Formation of HEL-specific 3A9 T cells was traced by flow cytometric
dent on Aire26.
To understand how Aire mutations affect the acquisition of immuno-
logical tolerance to organ-specific antigens, we intercrossed a targeted
Aire null mutation23 into a TCR transgenic strain that enables direct trac-
ing of organ-specific CD4 T cells14,27,28. The 3A9 TCR transgenic strain
carries rearranged TCRα and TCRβ genes, causing most developing
CD4+ helper T cells to express a single TCR with characterized high

Figure 2. Failure of thymic deletion in Aire–/– mice. Absolute numbers of


mature CD4 T cells in each thymus were calculated using two independent mea-
sures. Open bars indicate the mean numbers of CD4+CD8– 1G12hiCD69– T cells.
Filled bars indicate the mean numbers of CD4+CD8– 1G12hiCCL19 receptorhi T
cells. Error bars indicate standard deviation. Differences in mature cell numbers
were considered significant if t-test P values (two-sample assuming equal vari-
ances) were below 0.05, for both the number of CD4+CD8–1G12hiCD69– cells and
the number of CD4+CD8–1G12hiCCL19-Rhi cells. By this criterion, B10.Br TCR
mice showed no significant difference from Aire–/– TCR-insHEL mice, and both of
these groups showed substantially more mature cells than all other groups. Aire+/–
TCR-insHEL mice showed significantly higher levels of mature cells than B10.Br
TCR-insHEL mice, but not Aire+/+ TCR-insHEL mice. No other significant differ-
ences were observed.

www.nature.com/natureimmunology • april 2003 • volume 4 no 4 • nature immunology 351


A RTICLES

a b
© 2003 Nature Publishing Group http://www.nature.com/natureimmunology

Figure 3. Islet-specific T cells in the peripheral repertoire of Aire–/– mice.


Splenocytes were stained with 1G12 clonotypic antibody to TCR and CD4. (a) Repre-
sentative profiles from mice of the indicated genotypes. Numbers indicate the percent-
ages of splenocytes that fall within the indicated regions. (b) Bar graph showing the per-
centage of clonotypic TCRhiCD4+ splenocytes for mice of the indicated genotypes. Error
bars indicate standard deviation. Differences in cell numbers were considered significant
if t-test P values (two-sample assuming equal variances) were below 0.05. By this crite-
rion, B10.Br TCR mice showed substantially more clonotypic TCRhiCD4+ splenocytes
than all other genotypes, including Aire–/– TCR-insHEL mice. Aire–/– TCR-insHEL mice in
turn showed substantially more clonotypic TCRhi CD4+ splenocytes than B10.Br and
Aire+/+ TCR-insHEL mice. No other significant differences were observed.

staining with a 3A9 TCR clonotype-specific antibody, 1G1231, in the Production of CD4+CD25+ T cells
thymus of TCR-insHEL Aire–/– mice and in a range of littermate con- A longstanding alternative model for explaining tolerance to organ-
trols (Fig. 1). In the thymus of TCR-transgenic animals (lacking specific antigens is the concept of educating self-antigen–specific T
insHEL), T cells bearing the HEL-specific TCR were positively select- cells in the thymus to be exported as regulatory and/or suppressor cells
ed to mature as CD4+CD8– single-positive cells (Fig. 1a). The CD4+ T that prevent autoimmunity in the periphery. CD4+CD25+ cells in the
cells expressed high densities of the HEL-specific TCR on their sur- mouse have been suggested to have this property33–35. Therefore, the
face, and displayed the mature phenotype of down-regulated CD69 production of CD4+CD25+ putative regulatory T cells was also moni-
(activation/developmental marker) and up-regulated receptors (CCR7 tored during thymic development. In TCR transgenic mice without the
or CCR11) for the chemokine CCL19 (ELC)32 (Fig. 1a). Based on these insHEL transgene, only 2% of TCRhiCD4+CD8– T cells were CD25hi
markers, there was no difference in the positive selection of HEL-spe- (Table 1). In TCR-insHEL double-transgenic mice, the proportion of
cific T cells between Aire–/– and the background B10.Br (Aire+/+) TCR CD25+ T cells among TCRhiCD4+CD8– cells increased to 25%. This
transgenic animals (Supplementary Table 1). increase was completely mitigated in Aire–/– TCR-insHEL double-trans-
In the thymus of TCR-insHEL double-transgenic mice that were genic mice, with only 2% of TCRhiCD4+CD8– cells expressing high
either Aire+/+ littermates or B10.BR Aire wild-type animals, few CD4 amounts of CD25 (Table 1). When this subset of T cells was measured
single-positive cells were formed (Fig. 1b and Table 1, see as an absolute number, however, there was no significant difference
Supplementary Table 1 for more details) and of those a minority had between any of the groups. Thus, the increase in percentage of HEL-
matured to express high densities of HEL-specific TCR (Fig. 1b and specific CD4+25+ cells in Aire wild-type animals is secondary to the
Fig. 2), reflecting efficient T cell clonal deletion (negative selection). loss of HEL-specific CD4+25– cells.
Negative selection was also occurring in the immature CD4+8+ double-
positive population, so that the immature population was reduced to Thymus-intrinsic mechanism of Aire-mediated deletion
half that found in TCR animals (Table 1). The small number of CD4+ To determine the cellular requirement for Aire during negative selec-
T cells that mature in the TCR-insHEL thymus carry proportionately tion, chimeric mice were constructed with defective Aire in either the
reduced clonotype TCR and CD3, and ele-
vated CD5, on their cell surface (Fig. 1b).
We have previously shown that these resid- Table 2. The role of Aire in negative selection is intrinsic to the nonhemopoietic
ual cells are anergic based on poor prolifer- compartment
ative response to antigen or TCR Vβ anti- Donor mouse B10.Br TCR+ Aire–/– TCR+
body14. In contrast, in the thymus of Aire–/–
Recipient mouse B10.Br B10.Br HEL+ B10.Br B10.Br HEL+
TCR-insHEL mice, negative selection was
negligible in both the double-positive and Single-positive cells (CD4+CD8–) (%) 27 ± 2 7.8 ± 2 31 ± 8 10 ± 2
immature single-positive subsets. Mature CD4+CD8–1G12hi cells (%) 25 ± 4 4±3 29 ± 8 7±3
CD4+ T cells with high densities of HEL- CD4+CD8–1G12hi CD69– cells (%) 14 ± 2 1.2 ± 0.9 16 ± 5 2.4 ± 0.9
specific TCR, low CD69 and high expres-
sion of CCL19 receptor were produced in Donor mouse B10.Br TCR+
almost normal numbers, with normal cell- Recipient mouse B10.Br B10.Br HEL+ Aire–/– Aire–/– HEL+
surface expression of CD3 and CD5 (Fig.
1d, Fig. 2 and Table 1). This failure of Single-positive cells (CD4+CD8–) (%) 24 ± 2 9.6 ± 2 14 ± 4 13 ± 5
thymic negative selection resulted in CD4+CD8–1G12hi cells (%) 18.7 ± 2 5.1 ± 2 8.6 ± 5 8.7 ± 3
increased export of high-avidity, islet-HEL CD4+CD8–1G12hiCD69– cells (%) 5.9 ± 1 0.8 ± 0.4 2.4 ± 1 1.7 ± 0.6
specific T cells to the peripheral circulation Mean ± s.d.
(Table 1 and Fig. 3).

352 nature immunology • volume 4 no 4 • april 2003 • www.nature.com/natureimmunology


A RTICLES

hematopoietic or nonhematopoietic compartments. Analysis of thymic The defective thymic deletion of organ-specific CD4+ T cells in
composition after 6 weeks of reconstitution indicated that Aire–/– HEL- mice with an Aire mutation does not exclude the possibility that other
specific T cells were deleted normally in the thymus of Aire+/+ insHEL tolerance mechanisms may also depend on Aire. The answer will only
mice (Table 2 and Supplementary Table 2). In contrast, Aire+/+ HEL- be found with greater understanding of the mechanism by which Aire-
specific T cells were not deleted in Aire–/– insHEL mice, demonstrating dependent ectopic expression results in central tolerance. Although the
that the trait is intrinsic to the nonhematopoietic compartment (Table 2 mechanism is efficient for deleting 3A9 T cells, which recognize a
and Supplementary Table 2). These data correlate well with the dominant peptide epitope of HEL, TCR-transgenic T cells specific for
restriction of thymic Aire expression to the nonhematopoietic lin- a subdominant epitope of HEL are not deleted in the same insHEL
© 2003 Nature Publishing Group http://www.nature.com/natureimmunology

eages23,36, and the thymic-intrinsic induction of autoimmunity in transgenic line, even though these cells are still capable of responding
chimeric mice using Aire–/– mice as recipients for Aire+/+ bone marrow26. to antigen levels expressed in the periphery and islets (S. Hartley, M.
Noticeably, in the Aire–/– recipients without the insHEL transgene, pos- Neighbors and A. O’Garra, personal communication). Thymic dele-
itive selection of HEL-specific T cells seemed to be reduced in Aire–/– tion of high-affinity T cells recognizing dominant epitopes may be suf-
mice at this early time point, a trait which was not observed in ficient for tolerance, by reducing the autoreactive precursor frequency
nonchimeric Aire–/– TCR transgenic mice. This may, therefore, repre- to unresponsive levels. However, several other possibilities must be
sent a quantitative impediment in normal T cell maturation in Aire–/– considered. For example, the thymic induction of anergic T cells with
thymus, which is apparent during the wave of reconstitution, but not lower surface TCR-CD3 expression and elevated CD5, observed in the
once homeostasis is achieved. thymus of TCR-insHEL Aire wild-type mice, may be of functional
importance. Not only does anergy prevent autoreactive T cells from
Discussion responding39, but when precursor frequencies are reduced by thymic
We have demonstrated here the defect in thymic negative selection of deletion, residual anergic cells may be able to induce tolerance in the
high-avidity, organ-specific T cells in Aire–/– mice, which is one of the periphery40 via linked suppression (tolerogenic antigen-presenting
first instances in which the cellular mechanism for a human autoim- cells)41,42, competition for stimulatory cytokines43, or direct T cell–T
mune disease has been identified, and is the first evidence directly cell interactions10. In one TCR transgenic model, thymic expression of
connecting human autoimmune disease with defects in thymic dele- antigen induces the development of anergic autoreactive cells that cor-
tion. This result was unexpected because there is a body of evidence respond phenotypically and functionally to the subset of thymus-
supporting the view that tolerance to organ-specific antigens must dependent CD4+25+ regulatory T cells present in normal lymphocyte
depend, for the most part, on peripheral mechanisms acting on T cells repertoires33–35. However, in the model used here, Aire-dependent
after they have left the thymus, rather than on central tolerance dur- ectopic expression of insHEL did not alter the number of anti-HEL
ing T cell development, due to the low expression of organ-specific TCRhiCD4+CD25+ T cells in the thymus, although the efficient deletion
antigens in the thymus37. Moreover, we have previously considered of TCRhiCD25– T cells resulted in large shift in the ratio of autoreac-
that the thymic deletion to insHEL might be caused by circulating tive to CD4+CD25+ regulatory T cells in TCR-insHEL mice that was
HEL, which is cleaved from islet β cells and is present at nanomolar abrogated in Aire-deficient mice. Because CD4+CD25+ regulatory T
serum concentrations comparable to those of insulin itself14. However, cells seem to exist in a semi-anergic state44, Aire-dependent deletion
this idea is changing due to the unexpected discovery that the thymus and anergy may simultaneously lower the frequency of the most
is capable of promiscuous transcription of a wide range of ‘organ- strongly organ-reactive clones and enrich the remaining repertoire
specific’ genes17. Originally observed in transgenes with organ-spe- with organ-reactive anergic T cells that have a capacity to suppress the
cific promoters (including the rat insulin promoter used here16), arti- remaining naive T cells.
factual promoter ‘leakiness’ was excluded with the observation that The failure of thymic deletion provides a clearer understanding for
many organ-specific endogenous genes are also expressed in the thy- autoimmune polyendocrinopathy pathology by demonstrating that Aire
mus38. Ectopic expression has been proposed to reflect a specialized is essential for a highly efficient and specialized mechanism of thymic
physiological process, with rare PAE cells in the thymic medullary T cell deletion. This result is an important step for the field because it
epithelium expressing a range of peripheral antigens17. Anderson et connects human immunological tolerance with the large body of work
al.26 have recently shown that some ectopic mRNA expression is on thymic negative selection in transgenic mice. Bridging human
dependent on Aire, notably a defect in insulin mRNA transcription in autoimmunity and tolerance mechanisms in the mouse also has mech-
Aire–/– medullary stromal cells. These data, however, do not demon- anistic implications for more common and genetically complex autoim-
strate a biological role for Aire-dependent mRNAs, and the mecha- mune diseases. Comparable lesions in thymic deletion may be pro-
nism of any tolerance induced by Aire is not discussed. In the insHEL duced by genetic variants acting at various steps in the process of
transgenic mice studied here, membrane-bound HEL protein levels expression, presentation and recognition of organ-specific antigens by
were too low to detect on thymic stromal cells in Aire wild-type mice maturing thymocytes. For example, the type 1 diabetes susceptibility
(D. Gray and A.L., data not shown). Thus, it was not possible to deter- allele of the human insulin gene diminishes expression in the thy-
mine the direct effect of the absence of Aire on HEL expression in the mus45,46, and the diabetes-prone NOD mouse strain contains undeter-
thymus at the protein level. However, we conclude from the abroga- mined genetic lesions that result in a T cell intrinsic resistance to
tion of HEL-specific negative selection in Aire–/– mice that the rat thymic deletion27,47. Multiple sclerosis may also result from a defect in
insulin promoter drives Aire-dependent ectopic expression of self this process, as susceptibility to experimental autoimmune
antigen in a form capable of efficiently triggering T cell elimination, encephalomyelitis in the SJL/J mouse was found to correlate with the
despite the undetectable protein expression. Mixed bone-marrow thymic expression of a splice variant of the myelin-sheath protein, pro-
chimera experiments that dilute the precursor frequencies of TCR teolipid protein (PLP), which lacks the encephalitogenic epitope48,49.
transgenic cells indicate that the efficient deletion observed in TCR- Identifying other genes and molecules in the pathway of thymic cen-
insHEL thymus holds true at physiological precursor frequencies soring may therefore illuminate a common causality for a diverse range
(data not shown). of organ-specific autoimmune diseases.

www.nature.com/natureimmunology • april 2003 • volume 4 no 4 • nature immunology 353


A RTICLES

Methods 16. Jolicoeur, C., Hanahan, D. & Smith, K.M.T-cell tolerance toward a transgenic β-cell antigen and tran-
scription of endogenous pancreatic genes in thymus. Proc. Natl. Acad. Sci. USA 91, 6707–6711 (1994).
Mice. The 3A9 TCR transgenic28 and ILK-3 Ins-HEL transgenic14 mice produced in 17. Kyewski, B., Derbinski, J., Gotter, J. & Klein, L. Promiscuous gene expression and central T-cell toler-
C57BL/6J mice were backcrossed >7 generations to B10.Br/SgSnJ (JAX). (B6 × 129/Sv)F2 ance: more than meets the eye. Trends Immunol. 23, 364–371 (2002).
Aire+/– mice23 were backcrossed to TCR-insHEL B10.Br mice for two generations. 18. Betterle, C., Greggio, N.A. & Volpato, M. Clinical review 93: autoimmune polyglandular syndrome
Backcross offspring were typed for TCR, insHEL, H-214 and Aire23 by PCR, and H-2k Aire+/– type 1. J. Clin. Endocrinol. Metab. 83, 1049–1055 (1998).
mice of appropriate TCR-insHEL genotype were intercrossed to produce Aire–/– H-2k TCR- 19. Nagamine, K. et al. Positional cloning of the APECED gene. Nat. Genet. 17, 393–398 (1997).
20. The Finnish-German APECED Consortium. An autoimmune disease, APECED, caused by mutations
insHEL mice. Experimental mice were twice confirmed for genotype by PCR. Radiation
in a novel gene featuring two PHD-type zinc-finger domains. Nat. Genet. 17, 399–403 (1997).
chimeras were constructed as described14. Recipients were analyzed ∼6 weeks after recon- 21. Pitkanen, J. et al.The autoimmune regulator protein has transcriptional transactivating properties and
stitution. Animals were cared for and used in accordance with principles outlined by the interacts with the common coactivator CREB-binding protein. J. Biol. Chem. 275, 16802–16809
Australian National University Animal Experimentation Ethics Committee and the current (2000).
© 2003 Nature Publishing Group http://www.nature.com/natureimmunology

Australian Code of Practice for the Care and Use of Animals for Scientific Purposes. 22. Bjorses, P. et al. Mutations in the AIRE gene: effects on subcellular location and transactivation func-
tion of the autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy protein. Am. J. Hum.
Genet. 66, 378–392 (2000).
Flow cytometry. We analyzed 6- to 10-week-old mice (or 6 week post-reconstitution
23. Ramsey, C. et al. Aire deficient mice develop multiple features of APECED phenotype and show
chimeric mice) as described27,47 using the following antibodies: 1G12 anti-clonotype31 (gift altered immune response. Hum. Mol. Genet. 11, 397–409 (2002).
of E. Unanue and D. Peterson, Washington University, St. Louis, MO) culture supernatant 24. Heino, M. et al. Autoimmune regulator is expressed in the cells regulating immune tolerance in thy-
followed by rat anti-mouse IgG1 allo-phycocyanin; anti-CD8α-PerCP; anti-CD4-FITC or mus medulla. Biochem. Biophys. Res. Commun. 257, 821–825 (1999).
PE, anti-CD3-PE, anti-CD69-PE (all from BD PharMingen, San Jose, CA), anti-CD5-FITC 25. Bjorses, P. et al. Localization of the APECED protein in distinct nuclear structures. Hum. Mol. Genet.
and anti-CD25-PE (both from Caltag, Burlingame, CA). Staining for CCL19/ELC receptors 8, 259–266 (1999).
26. Anderson, M.S. et al. Projection of an immunological self-shadow within the thymus by the Aire pro-
was performed as described50 using CCL19-Fc fusion protein and goat polyclonal anti-
tein. Science 298, 1395–1401 (2002).
human IgG Fcγ-PE (Jackson Immunoresearch, West Grove, PA). 27. Lesage, S. et al. Failure to censor forbidden clone of CD4 T cells in autoimmune diabetes. J. Exp. Med.
196, 1175–1188 (2002).
Note: Supplementary information is available on the Nature Immunology website. 28. Ho,W.Y., Cooke, M.P., Goodnow, C.C. & Davis, M.M. Resting and anergic B cells are defective in
CD28-dependent costimulation of naive CD4+ T cells. J. Exp. Med. 179, 1539–1549 (1994).
Acknowledgments 29. Adelstein, S. et al. Induction of self-tolerance in T cells but not B cells of transgenic mice expressing
little self antigen. Science 251, 1223–1225 (1991).
We thank D. Gray and R. Boyd for thymic HEL measurement; J. Cyster, E. Unanue and D. 30. Akkaraju, S., Canaan, K. & Goodnow, C.C. Self-reactive B cells are not eliminated or inactivated by
Peterson for staining reagents; the staff of Medical Genome Centre for curating the autoantigen expressed on thyroid epithelial cells. J. Exp. Med. 186, 2005–2012 (1997).
mouse colony; and A. Murtagh, S. Ewing and S.Ward for genotyping.This work was sup- 31. Van Parijs, L., Peterson, D.A. & Abbas, A.K.The Fas/Fas ligand pathway and Bcl-2 regulate T cell
ported by a grant from the Juvenile Diabetes Research Foundation and NHMRC. responses to model self and foreign antigens. Immunity 8, 265–274 (1998).
32. Ueno,T. et al. Role for CCR7 ligands in the emigration of newly generated T lymphocytes from the
neonatal thymus. Immunity 16, 205–218 (2002).
Competing interests statement 33. Jordan, M.S. et al.Thymic selection of CD4+CD25+ regulatory T cells induced by an agonist self-pep-
tide. Nat. Immunol. 2, 301–306 (2001).
The authors declare that they have no competing financial interests.
34. Mason, D. Some quantitative aspects of T-cell repertoire selection: the requirement for regulatory T
Received 9 December 2002; accepted 11 February 2003. cells. Immunol. Rev. 182, 80–88 (2001).
35. Sakaguchi, S. Regulatory T cells: key controllers of immunologic self-tolerance. Cell 101, 455–458
1. Ohashi, P.S. et al. Ablation of “tolerance” and induction of diabetes by virus infection in viral antigen (2000).
transgenic mice. Cell 65, 305–317 (1991). 36. Heino, M. et al. RNA and protein expression of the murine autoimmune regulator gene (Aire) in
2. Katz, J.D.,Wang, B., Haskins, K., Benoist, C. & Mathis, D. Following a diabetogenic T cell from genesis normal, RelB-deficient and in NOD mouse. Eur. J. Immunol. 30, 1884–1893 (2000).
through pathogenesis. Cell 74, 1089–1100 (1993). 37. Stockinger, B.T lymphocyte tolerance: from thymic deletion to peripheral control mechanisms. Adv.
3. Goverman, J.Tolerance and autoimmunity in TCR transgenic mice specific for myelin basic protein. Immunol. 71, 229–265 (1999).
Immunol. Rev. 169, 147–159 (1999). 38. Hanahan, D. Peripheral-antigen-expressing cells in thymic medulla: factors in self-tolerance and
4. Scott, B. et al. A role for non-MHC genetic polymorphism in susceptibility to spontaneous autoim- autoimmunity. Curr. Opin. Immunol. 10, 656–662 (1998).
munity. Immunity 1, 73–83 (1994). 39. Schwartz, R.H.T cell anergy. Annu. Rev. Immunol. 21, 305–334 (2003).
5. Lafaille, J.J., Nagashima, K., Katsuki, M. & Tonegawa, S. High incidence of spontaneous autoimmune 40. Jooss, K., Gjata, B., Danos, O., von Boehmer, H. & Sarukhan, A. Regulatory function of in vivo aner-
encephalomyelitis in immunodeficient anti-myelin basic protein T cell receptor transgenic mice. Cell gized CD4(+) T cells. Proc. Natl. Acad. Sci. USA 98, 8738–8743 (2001).
78, 399–408 (1994). 41. Vendetti, S. et al. Anergic T cells inhibit the antigen-presenting function of dendritic cells. J. Immunol.
6. Waldner, H.,Whitters, M.J., Sobel, R.A., Collins, M. & Kuchroo,V.K. Fulminant spontaneous autoimmu- 165, 1175–1181 (2000).
nity of the central nervous system in mice transgenic for the myelin proteolipid protein-specific T 42. Taams, L.S. et al. Anergic T cells actively suppress T cell responses via the antigen-presenting cell. Eur.
cell receptor. Proc. Natl. Acad. Sci. USA 97, 3412–3417 (2000). J. Immunol. 28, 2902–2912 (1998).
7. Hernandez, J., Aung, S., Redmond,W.L. & Sherman, L.A. Phenotypic and functional analysis of CD8(+) 43. Lombardi, G., Sidhu, S., Batchelor, R. & Lechler, R. Anergic T cells as suppressor cells in vitro. Science
T cells undergoing peripheral deletion in response to cross-presentation of self-antigen. J. Exp. Med. 264, 1587–1589 (1994).
194, 707–717 (2001). 44. Sakaguchi, S. et al. Immunologic tolerance maintained by CD25+ CD4+ regulatory T cells: their com-
8. Lambolez, F., Jooss, K.,Vasseur, F. & Sarukhan, A.Tolerance induction to self antigens by peripheral mon role in controlling autoimmunity, tumor immunity, and transplantation tolerance. Immunol. Rev.
dendritic cells. Eur. J. Immunol. 32, 2588–2597 (2002). 182, 18–32 (2001).
9. Greenwald, R.J., Boussiotis,V.A., Lorsbach, R.B., Abbas, A.K. & Sharpe, A.H. CTLA-4 regulates induc- 45. Vafiadis, P. et al. Insulin expression in human thymus is modulated by INS VNTR alleles at the IDDM2
tion of anergy in vivo. Immunity 14, 145–155 (2001). locus. Nat. Genet. 15, 289–292 (1997).
10. Shevach, E. CD4+CD25+ suppressor T cells: more questions than answers. Nat. Rev. Immunol. 2, 46. Pugliese, A. et al.The insulin gene is transcribed in the human thymus and transcription levels corre-
389–400 (2002). lated with allelic variation at the INS VNTR-IDDM2 susceptibility locus for type 1 diabetes. Nat.
11. Le Douarin, N. et al. Evidence for a thymus-dependent form of tolerance that is not based on elimi- Genet. 15, 293–297 (1997).
nation or anergy of reactive T cells. Immunol. Rev. 149, 35–53 (1996). 47. Kishimoto, H. & Sprent, J. A defect in central tolerance in NOD mice. Nat. Immunol. 2, 1025–1031
12. Hammerling, G.J. et al. Non-deletional mechanisms of peripheral and central tolerance: studies with (2001).
transgenic mice with tissue-specific expression of a foreign MHC class I antigen. Immunol. Rev. 122, 48. Klein, L., Klugmann, M., Nave, K.A.,Tuohy,V.K. & Kyewski, B. Shaping of the autoreactive T-cell reper-
47–67 (1991). toire by a splice variant of self protein expressed in thymic epithelial cells. Nat. Med. 6, 56–61
13. Hoffmann, M.W., Heath,W.R., Ruschmeyer, D. & Miller, J.F. Deletion of high-avidity T cells by thymic (2000).
epithelium. Proc. Natl. Acad. Sci. USA 92, 9851–9855 (1995). 49. Anderson, A.C. et al. High frequency of autoreactive myelin proteolipid protein-specific T cells in the
14. Akkaraju, S. et al. A range of CD4 T cell tolerance: partial inactivation to organ-specific antigen periphery of naive mice: mechanisms of selection of the self-reactive repertoire. J. Exp. Med. 191,
allows nondestructive thyroiditis or insulitis. Immunity 7, 255–271 (1997). 761–770 (2000).
15. Klein, L., Klein,T., Ruther, U. & Kyewski, B. CD4 T cell tolerance to human C-reactive protein, an 50. Reif, K. et al. Balanced responsiveness to chemoattractants from adjacent zones determines B-cell
inducible serum protein, is mediated by medullary thymic epithelium. J. Exp. Med. 188, 5–16 (1998). position. Nature 416, 94–99 (2002).

354 nature immunology • volume 4 no 4 • april 2003 • www.nature.com/natureimmunology

You might also like