You are on page 1of 15

REVIEWS

HLA variation and disease


Calliope A. Dendrou1, Jan Petersen2,3, Jamie Rossjohn2–4 and Lars Fugger5,6
Abstract | Fifty years since the first description of an association between HLA and human
disease, HLA molecules have proven to be central to physiology, protective immunity and
deleterious, disease-causing autoimmune reactivity. Technological advances have enabled
pivotal progress in the determination of the molecular mechanisms that underpin the association
between HLA genetics and functional outcome. Here, we review our current understanding of
HLA molecules as the fundamental platform for immune surveillance and responsiveness in
health and disease. We evaluate the scope for personalized antigen-specific disease prevention,
whereby harnessing HLA–ligand interactions for clinical benefit is becoming a realistic prospect.

Linkage disequilibrium HLA gene variation was first linked to human disease More than 15,000 different classical HLA class I and
The nonrandom association of with the discovery of the association between HLA‑B II alleles have been identified4. The diversity of HLA
alleles at different loci, for and Hodgkin lymphoma1. Since then, the MHC has molecules expressed in humans at the population level
example, owing to close been established as the region of the genome that is likely to maximize the probability that at least some
physical proximity within a
genomic region.
is associated with the greatest number of human individuals within the general population can mount an
diseases 2. The majority of these diseases have an immune attack against an emerging infection and survive.
Gene conversion immuno­logical component, which is consistent with Genetically, this diversity may arise partly due to point
The process by which one the enrichment for key immune genes within the MHC mutations but also more frequently due to mecha­nisms
allele is converted to another
region. The MHC is divided into three subclasses: the such as gene conversion2. Much of the poly­morphism in
by mismatch repair
mechanisms.
class I region, which includes the classical, highly poly­ HLA genes leads to nonsynonymous amino acid changes
morphic HLA‑A, HLA‑B and HLA‑C genes, as well as in the peptide-binding groove of HLA molecules, thus
Heterozygote advantage the nonclassical HLA‑E, HLA‑F and HLA‑G genes, indicating a strong selection pressure on the peptide-­
The increased relative fitness of which exhibit limited polymorphism; the class  II binding groove and the importance of the HLA–peptide
an organism conferred by
having two different forms of a
region, which includes the HLA‑DPA1, HLA‑DPB1, interaction. Two main mechanisms of balancing selection
genetic variant, as opposed to HLA‑DQA1, HLA‑DQA2, HLA‑DQB1, HLA‑DQB2, have been postulated that favour the maintenance of HLA
having two identical copies of HLA-DRA, HLA‑DRB1, HLA‑DRB2, HLA‑DRB3, diversity in a population over time: heterozygote advan-
either of the two forms. HLA‑DRB4 and HLA‑DRB5 genes as well as less var­ tage and frequency-dependent selection5. The potential of
iable genes involved in antigen processing and pres­ pathogens to drive HLA selection is suggested through
Frequency-dependent
selection entation; and the class III region, which contains genes investigations of emerging infectious diseases, such as
An evolutionary process implicated in inflammatory responses, leukocyte matu­ HIV; in HIV infection, HLA class I allele heterozygosity,
whereby fitness of a given ration and the complement cascade. Nonpolymorphic as opposed to homozygosity, correlates with delayed dis­
phenotype depends on its antigen-presenting molecules that are encoded by genes ease progression6. However, resistance to infection may
frequency relative to other
phenotypes in a study
outside of the MHC locus (for example, CD1a–d and not be the only driver of HLA polymorphism, as HLA
population. Positive selection MHC class-I‑related protein) have been reviewed else­ molecules are implicated in reproductive fitness via the
will occur if the fitness of the where3 and are not the main focus of this Review. The regulation of maternal and fetal survival7.
phenotype increases as its MHC locus has a dense clustering of immune-­relevant In an individual who is heterozygous at each of the
frequency increases, whereas
genes that can show extreme polymorphism and can six classical class I or II HLA loci, it has been estimated
negative selection will occur if
the fitness decreases as the
be in strong linkage disequilibrium; this complexity has that an antigen-presenting cell (APC) could theo­retically
frequency of the phenotype complicated the determination of the exact genes and present over 1012 different peptides8. The specific recog­
increases. alleles that are responsible for signals of disease associ­ nition of HLA–peptide combinations is mediated by αβ T
ation in the region2. However, the classical HLA class I cell receptors (TCRs) on CD8+ T cells, which bind class I
and II genes are often thought to drive associations molecules, and on CD4+ T cells, which bind class II mole­
Correspondence to L.F. and J.R. 
with disease. This is in keeping with the crucial role cules. The TCR displays substantial sequence hetero­
lars.fugger@imm.ox.ac.uk; that HLA class I and II molecules have in presenting a geneity that arises as different variable (V), diversity (D)
jamie.rossjohn@monash.edu vast array of antigenic peptides to T cells, thus enabling and joining (J) gene segments come together through
doi:10.1038/nri.2017.143 the immune system to discriminate between self and somatic, convergent recombination; additional variation
Published online 2 Jan 2018 non-self. is introduced by the semi-random insertion or deletion of

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 1


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Author addresses
Figure 1 | HLA-dependent molecular mechanisms of ▶
peptide and T cell receptor binding. a | Conventional
1
Nuffield Department of Medicine, The Wellcome Centre for Human Genetics, docking. Central docking geometry with the T cell receptor
University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK. (TCR) α-chains and β-chains positioned diagonally with
2
Australian Research Council Centre of Excellence for Advanced Molecular Imaging, respect to the peptide, thus maximizing the coverage of the
Monash University, Wellington Road, Clayton, Victoria 3800, Australia. peptide–HLA surface and allowing the TCR to access
3
Infection and Immunity Programme and Department of Biochemistry and Molecular exposed peptide side chains. b | Alternate docking.
Biology, Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, Unconventional amino-terminal docking geometry with
Victoria 3800, Australia. α-chains and β-chains positioned perpendicularly with
4
Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, respect to the peptide axis, thus limiting coverage of the
Cardiff CF14 4XN, UK. peptide–HLA surface and preventing interactions between
5
Danish National Research Foundation Centre PERSIMUNE, Rigshospitalet, the TCR and the carboxy‑terminal portion of the peptide.
University of Copenhagen, Copenhagen DK-2100, Denmark. Top row of parts a and b depicts the interface viewed along
6
Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, the TCR–peptide–HLA axis; bottom row of parts a and b,
Division of Clinical Neurology and Medical Research Council Human Immunology Unit, and parts c–i depict a cross section of the interface along
Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, the peptide backbone. c | TCR stabilization of weak
University of Oxford, Headley Way, Oxford OX3 9DS, UK. peptide–HLA complexes. Left: unstable peptide–HLA
complex with suboptimal peptide anchor residues.
Right: TCR binding to the peptide–HLA stabilizes the weak
nucleotides at segment junctions9. As a result, each TCR complex interactions. d | Altered register. Polymorphic
chain bears three highly variable complementarity deter­ differences between HLA‑DR1 and HLA‑DR15 result in
mining region (CDR) loops: CDR1 and CDR2 diversity different anchor residues being used for α3135–145 peptide
is germline-encoded, whereas the CDR3 loop is hyper­ presentation, resulting in vastly different antigenic surfaces.
variable, as it is encoded by the sequences generated by e | Hotspot binding and molecular mimicry. TCRs that form
nucleotide insertion and deletion. In addition to binding focal interactions with only a limited portion of the cognate
TCRs (the focus of this Review), HLA class I molecules peptide–MHC complex (left) tend to recognize mimic
peptides (right) that differ in their primary sequence.
can also bind killer cell immunoglobulin-like receptors
f | Post-translational modification (PTM)-altered HLA
(KIRs) and C‑type lectin-like CD94/NKG2 family of binding. PTMs can substantially change peptide binding to
receptors found on natural killer (NK) cells10 as well as the HLA and expose new features to the TCR. Left: a native
leukocyte immunoglobulin-like receptors (LILRs) found peptide is unable to bind the HLA. Right: the peptide
on APCs and several other cell types11. These interactions carrying PTMs is presented by the HLA because of its
are thought to be less peptide-specific than HLA–TCR changed chemistry and exposes a modified residue for TCR
binding, although there is evidence that they can be recognition. g | PTM-altered antigen processing. Left: an
tuned in a peptide-dependent fashion11–13. antigenic peptide is cleaved (red dashes) into fragments too
The specificity of HLA–peptide–TCR tripartite inter­ short for HLA presentation. Right: PTM of the same peptide
actions is fundamental in enabling the adaptive immune obscures a protease site, resulting in productive antigen
processing for HLA presentation. h | HLA presentation of
system to mount an efficient and appropriate response
hybrid peptides. Top: two peptides with suboptimal anchor
to counteract infection and malignancy while maintain­ residues are poorly bound by the HLA. Bottom: the hybrid
ing self tolerance and preventing autoimmune disease. peptide has enhanced anchor residues for HLA binding and
Understanding the molecular principles that govern provides novel features for TCR recognition. i | Stability and
these interactions may give mechanistic insight into the peptide repertoire. The comparatively wide and shallow
role of HLA in driving and protecting against immuno­ peptide-binding groove of HLA‑C*05 enables presentation
pathology; this presents an ongoing biomedical research of peptides with large, aromatic anchor residues, whereas
challenge but also holds much therapeutic promise. the deep and narrow peptide-binding groove of HLA‑C*07
In this Review, we consider our current understand­ selects for smaller anchor residues. This difference
ing of HLA function in immune-mediated diseases contributes to HLA‑C*05 forming more stable complexes,
which results in higher expression levels on the cell surface
with a particular emphasis on autoimmune conditions.
of HLA‑C*05 compared with HLA‑C*07.
Following this, we discuss how technological progress
is facilitating the study of HLA biology, and we explore
the prospects for developing and using antigen-specific complex structures has suggested that there are certain
strategies for disease prevention and therapy. principles that govern HLA restriction and TCR docking
geometry, but that they are malleable; instead of a conven­
HLA interactions in disease tional, central TCR docking geometry (FIG. 1a), a variety
The structural determination of disease-relevant of molecular mechanisms that affect HLA–peptide–TCR
peptide–HLA and HLA–peptide–TCR complexes is interactions are implicated in disease14 (FIG. 1b–i). A still
crucial for the elucidation of the molecular mechanisms open question is whether any of these mechanisms are
responsible for the development of specific protective shared between conditions with common aetiological
HLA restriction immunity against pathogens, as well as for the deleter­ features. For example, HLA–peptide–TCR binding has
A property of T cells whereby a ious T cell reactivity that promotes disease. One model been of particular interest in the context of different
given T cell receptor will proposes that the HLA–peptide–TCR interactions that autoimmune diseases. A common feature of these dis­
recognize and respond to an
antigen only when it is
underpin self and non-self discrimination are guided by eases is tissue damage facilitated by autoreactive T cells
presented by a particular HLA a set of rules that are perturbed in immunopathology. that escape both negative selection in the thymus and
molecule. The study of more than 50 distinct HLA–peptide–TCR peripheral tolerance mechanisms. However, it remains

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a Conventional docking b Alternate docking c TCR stabilization of weak peptide–HLA complexes

TCRβ Peptide
TCRβ

C C
N N
HLA
TCRα TCRα
Peptide

HLA

d Altered register e Hotspot binding and molecular mimicry


Focal interaction
αβTCR

α3–HLA-DR1 α3–HLA-DR15 Cognate peptide–MHC Mimic peptide

f PTM-altered HLA binding g PTM-altered antigen processing


Site of peptide
cleavage

h HLA presentation of hybrid peptides i Stability and peptide repertoire

HLA-C*05 HLA-C*07
Hybrid peptide

Native
Mimic peptide
peptide PTM residues
residues
residues

to be determined whether autoreactive T cells are directly Alternate docking. The Ob.1A12 TCR, which was orig­
generated and activated by the mechanisms that are inally isolated from CD4+ Nature
T cellsReviews
from a |patient with
Immunology
described below, such as atypical HLA–peptide–TCR relapsing–remitting multiple sclerosis, recognizes resi­
binding orientation, low-affinity peptide binding that dues 85–99 of the immunodominant peptide of myelin
facilitates thymic escape, TCR-mediated stabilization of basic protein (MBP), which is presented by the dis­
weak peptide–HLA interaction and presentation of pep­ ease-associated HLA‑DR15. The pathogenic potential of
tides in a different binding register. Other mechanisms the Ob.1A12 TCR in promoting central nervous system
that may generate and activate autoreactive T cells are (CNS) disease has been demonstrated in transgenic mice
likely to be driven by epitope variation, including molec­ expressing Ob.1A12 TCR and HLA‑DR15 and immu­
ular mimicry, post-translational epitope modification nized with the MBP peptide15. At the molecular level, the
and the generation of hybrid peptides, and processes that Ob.1A12 TCR displays an off-centre binding topology
regulate HLA expression and stability are also emerging (FIG. 1b), whereby it docks over the amino‑terminal side
as being increasingly relevant (FIG. 1b–i). of the peptide–HLA platform16. This alternate docking

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 3


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

topology was associated with reduced affinity of the peptide-specific TCRs22 and probably exerts its com­
TCR for the peptide–HLA complex and has an uncon­ pensatory effect through the maximal binding of MBP.
ventional geometry that may have allowed autoreactive As the HLA‑DR4‑MBP peptide interaction is highly
T cell evasion of thymic negative selection, although unstable, a low autoantigen density in the thymus could
still maintaining the capacity of the TCR to stimulate an have enabled MS2‑3C8‑TCR-bearing T cells to receive
inflammatory response in the periphery (TABLE 1). a weak signal for positive selection that would not reach
the required threshold for negative selection. Once in
Low-affinity-mediated thymic escape. The 1E6 TCR, the periphery, MS2‑3C8 TCR+ cells could then be acti­
which was isolated from CD8+ T cells from a newly vated in response to higher antigen densities, potentially
diagnosed patient with type 1 diabetes (T1D), medi­ triggering an autoimmune response against the CNS.
ates pancreatic islet β-cell killing by recognizing amino
acids 15–24 of the preproinsulin (PPI) signal peptide Altered register. The peptide binding register refers to
presented by disease-associated HLA‑A*02:01. The 1E6 the ~9‑mer window of a peptide that sits directly within
TCR binds this peptide–HLA complex in a conventional the peptide-binding groove at a given time. Alterations
fashion, which is a characteristic of the majority of HLA in this register, whereby the same peptide binds a
class-I‑restricted TCRs studied to date17. However, the peptide-­binding groove utilizing a different 9‑mer win­
affinity of the 1E6 TCR for the peptide–HLA complex dow, can potentially have a differential impact on the
is very low and may be below the threshold for thymic generation of autoreactive T cells. Diabetogenic T cells
nega­tive selection induction, but it is nevertheless suf­ from nonobese diabetic (NOD) mice recognize an insu­
ficient to trigger β-cell killing (TABLE 1). Given the low-­
lin B‑chain peptide, which is bound to the MHC class II
affinity mode of binding of the TCR, the level of the I-Ag7 molecule in a low-affinity register 23, thus demon­
PPI peptide presented in the thymus may be related strating the potential importance of the peptide bind­
to the escape from negative selection. PPI expression in ing register. Similar results were seen in patients with
the thymus is under the influence of a variable number T1D, and there is also some evidence in these patients of
of tandem repeats (VNTRs) in the 5ʹ regulatory region of potential priming of diabetogenic T cells through molec­
the INS gene. The presence of few VNTRs is a strong risk ular mimicry of the self peptide (discussed below) with
factor for T1D and is associated with lower levels of PPI- microbial antigens24.
encoding mRNA transcripts in the thymus18. Notably, In addition to the potential role of the peptide
the patient from whom the 1E6 TCR was derived was binding register in increasing the risk of auto­immune
homozygous for VNTR alleles with few repeats in the disease, another fundamental question is its rele­
INS gene. In the periphery, the escapee CD8+ 1E6‑TCR- vance to disease resistance. This was the focus of a
bearing cells may be primed by microbial-derived recent study investigating the mechanisms by which
antigens, particularly as this TCR focuses on only two HLA‑DR15 and HLA‑DR1 confer respective risk of
residues within the PPI peptide, suggesting a higher or dominant protection against the renal autoimmune
chance of cross recognition. This model is supported by condition Goodpasture disease25. Autoreactive T cells
the finding that the 1E6 TCR can bind with a high affin­ specific for the α3 chain of type IV collagen (α3135–145)
ity to a peptide from the human pathogen Clostridium were expanded in patients with Goodpasture disease,
asparagi­forme19. In the pancreas, the pro-­inflammatory and immunization of HLA‑DR15 transgenic mice
environment that accompanies T1D development with the α3135–145 self peptide led to the infiltration
induces islet β-cells to upregulate HLA class I expres­ of peptide-­specific T cells into the kidney, resulting
sion, thus enabling the autoreactive T cells to exert theirin disease. Compared with HLA‑DR15, HLA‑DR1
cytotoxic function17. has a distinct peptide repertoire and binding prefer­
ences and presents the α3135–145 epitope in a different
T cell receptor stabilization of weak peptide–HLA binding register (FIG. 1d; TABLE 1). This altered regis­
complexes. Intriguingly, there is also evidence that ter promotes the generation of peptide-specific CD4+
auto­reactive TCRs can bind self peptide–HLA com­ T cells with a predominantly forkhead box protein P3
plexes with a conventional binding topology and (FOXP3)+ regulatory phenotype in both HLA‑DR1+
a high affinity 20. The MS2‑3C8 TCR was isolated and HLA‑DR15+HLA‑DR1+ transgenic mice, which
from CD4 + T  cells from a patient with relapsing–­ were resistant to disease. Consistent with this, healthy
remitting multiple sclerosis and binds an MBP pep­ human donors who were HLA‑DR15+ or HLA‑DR1+
tide presented by HLA‑DR4. The pathogenic potential displayed altered α3135–145-specific TCR usage and had
of this TCR has been demonstrated through the use of peptide-specific T cells that primarily showed either an
HLA‑DR4‑transgenic mice that developed CNS disease effector or a regulatory pheno­type, respectively 25. Thus,
via adoptive transfer of MS2‑3C8 TCR+CD4+ T cells in one mechanistic basis for dominant protective effects
the absence of antigen administration21. Structural ana­ of HLA alleles in autoimmunity, via the effect on the
lyses have shown that although the MBP peptide binds generation of CD4+CD25+ regulatory T (Treg) cells, has
HLA‑DR4 only weakly and is unstably accommodated been revealed.
in the peptide-binding cleft, the MS2‑3C8 TCR binds
the complex tightly and compensates for the weak MBP ‘Hotspot’ molecular mimicry. In the ‘hotspot’ molecular
peptide–HLA interaction20 (FIG. 1c; TABLE 1). The affinity mimicry model, autoreactive TCRs with a highly focused
of this TCR is in the range of that observed for pathogen footprint, which predominantly binds only a small area

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 1 | Autoimmune disease–HLA associations for which molecular mechanisms of action have been identified
HLA allomorph (effect Autoimmune Molecular mechanism of action Refs
on disease) disease
Alternate docking
HLA‑DR15 (risk) Multiple sclerosis • Alternate docking of a TCR may allow MBP peptide-specific T cells to escape thymic 15,16
selection
• In the periphery, such cells may be cross-reactive with microbial peptides
Low-affinity-mediated thymic escape
HLA‑A*02:01 (risk) T1D • Low affinity of a TCR for preproinsulin signal peptide–HLA may allow autoreactive 17–19
T cells to escape thymic selection
• This mechanism may depend on the thymic expression level of the autoantigen
• In the periphery, these cells may bind microbial peptides with a high affinity
TCR stabilization of weak peptide–HLA complexes
HLA‑DR4 (risk) Multiple sclerosis • Weak MBP peptide–HLA‑DR4 interaction is stabilized by binding of a 20–22
patient-derived TCR
• Low autoantigen density in the thymus may enable escape of autoreactive T cells
from negative selection
• Higher autoantigen density in the periphery could trigger an autoreactive response
Altered register
HLA‑DQ2 and HLA‑DQ8 T1D • Insulin B‑chain peptide binds HLA‑DQ2 and HLA-DQ8 with a low-affinity peptide 23,24
(risk) register
• Low affinity may allow autoreactive T cells to escape thymic selection
• In the periphery, autoreactive T cells may be cross-reactive with microbial peptides
via molecular mimicry
HLA‑DR15 (risk) Goodpasture disease • HLA‑DR15 and HLA‑DR1 both bind an α3‑chain of type IV collagen peptide 25
HLA‑DR1 (dominant (α3135–145) but with a different binding register
protection) • When HLA‑DR1 is present, this promotes the generation of autoantigen-specific
regulatory T cells as opposed to effectors
‘Hotspot’ molecular mimicry
HLA‑DR15 (risk) Multiple sclerosis • The probability of aberrant, off-target TCR reactivity induced by pathogen-derived 16,
peptides is increased for autoreactive TCRs with a highly focused footprint that 26–28,
predominantly binds only a small area of peptide 32
• Patient TCRs have been identified that cross-react with HLA‑DR15‑restricted MBP
and Escherichia coli or EBV-derived peptides
Post-translational modification
HLA‑DQ2.5 and HLA‑DQ8 Coeliac disease • HLA‑DQ2.5 and HLA-DQ8 present deamidated gliadins with a high kinetic stability, 38–41
(risk) leading to sustained antigen presentation that may promote a pathogenic T cell
response
HLA‑DR4 (risk) Rheumatoid arthritis • Citrullination of autoantigens (for example, vimentin) facilitates binding to HLA‑DR4 34,35
• Citrullination can also modify peptide cleavage, enabling retention of autoantigens
HLA‑DR4 (risk) T1D • Vicinal disulfide bond creation in an insulin A‑chain peptide presented by HLA‑DR4 37
is needed for recognition by a patient-derived autoreactive TCR
Hybrid peptides
HLA‑DQ8 (risk) T1D • Hybrid proinsulin peptides present in pancreatic β-cells may drive a breakdown in 51
immune tolerance
Regulation of HLA expression
MHC risk variants in distal Systemic lupus • Risk variants alter the binding of the IRF4 and CTCF factors that regulate the 52
intergenic XL9 regulatory erythematosus transcription of HLA‑DRB1, HLA‑DQA1 and HLA‑DQB1, resulting in a 2.5‑fold
element increase in the levels of the HLA‑DR and HLA‑DQ proteins implicated in lupus
Highly expressed HLA‑C Crohn’s disease • HLA‑C allotypes with a high expression correlate with Crohn’s disease risk 53
allotypes (risk) • This expression is partly regulated by the microRNA miR‑148a, which is itself
subject to regulation by genetic variation
HLA stability
HLA‑DQ2 and HLA‑DQ8 T1D, coeliac disease • HLA‑DQ2 and HLA-DQ8 instability could confer risk by allowing escape of 71–76
(risk) autoreactive T cells from thymic negative selection but also by the formation of
T1D, autoimmune
weak peptide–HLA complexes in the periphery with a range of autoantigens
HLA‑DQ6 (protection) polyglandular
• By a converse mechanism, the stability of HLA‑DQ6 may confer protection
syndrome, IgA
against autoimmune disease; despite such a mechanism, HLA‑DQ6 confers risk of
deficiency
narcolepsy
CTCF, transcriptional regulator CTCF; EBV, Epstein–Barr virus; IgA, immunoglobulin A; IRF4, interferon regulatory factor 4; MBP, myelin basic protein;
T1D, type 1 diabetes; TCR, T cell receptor.

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 5


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

of peptide, increase the probability of aberrant, off-­target present gluten-derived peptides (gliadins) that have been
TCR reactivity that is induced by pathogen-­derived deamidated by trans­glutaminase 2 (REF. 38). Gliadins are
peptides (FIG. 1e). For example, the alternate docking bound to HLA‑DQ2.5 with a high kinetic stability that
strategy used by the Ob.1A12 TCR when binding MBP leads to their sustained presentation, which is a require­
peptide16 was also observed when this TCR recognizes ment for the generation of the pathogenic T cell response.
a cross-reactive peptide from Escherichia coli 26 (TABLE 1). By contrast, HLA‑DQ2.2 confers a low risk of coeliac dis­
Importantly, this finding provided the first evidence that ease, and its geometry is such that gliadins cannot be sta­
atypical binding orientations are not only a feature of bly accommodated in the peptide-­binding cleft 39,40. The
aberrant reactivity but are also implicated in responses disease-associated HLA‑DQ8 lacks an aspartic acid resi­
to pathogen and that hotspot molecular mimicry may be due at position 57β, which creates a positively charged P9
a trigger for multiple sclerosis26. Similarly, the Hy.1B11 pocket with a preference for nega­tively charged peptides41.
TCR, which was derived from a patient with multiple The autoimmune disease rele­vance of the HLA‑DQ8
sclerosis, binds the MBP85–99 peptide, but only one of the polymorphic residue 57β was first identified for T1D42.
CDR3 loops makes contact with the peptide, thereby Furthermore, autoantigenic glutamic acid decarboxy­
limiting the specificity of the TCR to only a few resi­ lase 65 (GAD65) peptides bind with high affinity to the
dues within the peptide27. As the Hy.1B11 TCR relies on NOD mouse MHC class II I-Ag7 molecule, which also
only a few key residues, it can be activated not only by lacks this aspartic acid43. In the context of coeliac dis­
MBP but also by peptides from several different viruses, ease, the absence of this amino acid may better accom­
although these viral peptides show a low similarity only modate deamidated gliadins and may also promote the
to the MBP85–99 peptide28. The potential importance of recruitment of cross-reactive TCRs that carry a negative
molecular mimicry between self and microbial peptides signature charge in CDR3β, which can respond to both
in driving autoimmune disease is also supported by epi­ the modified and unmodified gluten peptides41 (TABLE 1).
demiological investigation and shared genetic factors. A shared feature of different coeliac disease-relevant
For example, for patients with multiple sclerosis, there peptide–HLA‑DQ complexes is their bias towards inter­
is an increased comorbidity with Epstein–Barr virus acting with certain TCRs; an arginine residue within the
(EBV)-dependent infectious mononucleosis29, and hav­ CDR3 loop of the TCRs studied is thought to anchor
ing HLA‑DRB1*15:01 increases the risk of developing the interaction with HLA‑DQ2.5–deamidated gliadin
both conditions30,31; these findings present the possibil­ and HLA‑DQ8–deamidated gliadin complexes44,45. These
ity that autoreactive T cells could in some instances be HLA–deamidated gliadin–TCR interactions may be at
activated by EBV antigens. Although numerous differ­ the centre of a pathological T cell–B cell cooperation34.
ent mechanisms may act to trigger multiple sclerosis, a The transglutaminase 2 enzyme that mediates gliadin
patient-derived TCR has been identified that recognizes deamidation also serves as a target for auto­antibodies
both HLA‑DRB1*15:01‑restricted MBP and an EBV in coeliac disease46. Thus, B cells with the capacity to
peptide presented by HLA‑DRB5*01:01 (TABLE 1), which generate such autoantibodies would initially bind trans­
is encoded by an allele in high linkage disequilibrium glutaminase 2–gliadin complexes via their B cell recep­
with the HLA‑DRB1*15:01 allele32. This supports the tors (BCRs), thereby mediating BCR-dependent gliadin
model of molecular mimicry acting to increase the risk internalization and presentation by HLA‑DQ2.5. CD4+
of autoimmune disease. T cells that become activated by the interaction of their
TCR with HLA‑DQ2.5–gliadin would then provide
Post-translational modification. The HLA-mediated help to the B cells, leading to the secretion of the auto­
presentation of neoantigens has been proposed as another antibodies that can bind transglutaminase 2, affinity
mechanism by which T cells could mount an attack against matu­ration, further gliadin uptake and presentation, and
self tissue (FIG. 1f). Post-translational modification (PTM) an ensuing boost of the T cell response34.
can generate such neoantigens through deamidation and A similar mechanism may also occur in rheumatoid
citrullination, processes that have been implicated in sev­ arthritis. CD4+ T cells that respond to citrullinated pep­
eral autoimmune diseases33–35. In addition, neoantigens tides presented by disease-associated HLA‑DR mole­
can be generated through glycosylation and phosphory­ cules that are present on citrullinated-protein-specific
lation36. Interestingly, post-translational vicinal disulfide B cells can then provide help to these B cells; the B cells
bond creation between adjacent cysteine residues was can then undergo class switching to secrete antibodies
found to be required for the TCR-mediated recognition specific for anti-citrullinated protein47 and may in turn
of an insulin A‑chain peptide in T1D, although this modi­ further stimulate the T cells. Notably, citrullination facil­
fication does not alter the binding of the peptide to HLA37. itates the binding of autoantigens to the rheumatoid-
Correlating processes of PTM to disease-­associated anti­ arthritis-­associated HLA‑DR4 protein. In addition, when
genic diversity is still a nascent research area. In the con­ the rheumatoid arthritis-associated protein vimentin
Neoantigens
Non-germline-encoded (and text of autoimmune disease, an assumption is made by becomes citrullinated, it also modifies protease-mediated
thus non-inherited) antigens researchers that PTM occurs in the periphery but not cleavage so that self peptides derived from vimentin tend
that may arise because of in the thymus during central tolerance establishment; to be retained rather than degraded35 (FIG. 1 g; TABLE 1).
somatic mutation (as in cancer) however, this concept requires formal testing.
or other processes such as
post-translational modification
The impact of deamidation has been revealed through Hybrid peptides. Another mechanism for neo­antigen
and splicing together of studies of T cell recognition in coeliac disease, where the generation is through the creation of hybrid self
peptides. disease-associated HLA‑DQ2.5 and HLA‑DQ8 molecules peptides in peripheral tissues (FIG. 1 h). Proteasomal

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

degradation of intracellular proteins to produce the class II transcription56. However, the interplay between
peptides typically presented by HLA class I mole­ EBV immune evasion mechanisms and viral latency and
cules can promote the generation of spliced epitopes. the link between EBV and multiple sclerosis remain to
Although this was once thought to be a rare occurrence, be determined.
it has now been demonstrated that up to one-third of The relevance of HLA expression levels in transplan­
the HLA class I peptidome comprises spliced peptides, tation has also been considered fairly recently. HLA‑C
thus indicating a greater diversity of presented self anti­ expression levels can influence permissivity to donor
gens or pathogen-derived antigens than previously mismatch and hence mortality after transplantation
appreciated48. Consequently, it is not surprising that and graft-versus-host disease (GVHD)57. Furthermore,
viral immune evasion mechanisms include resistance HLA‑DPB1‑mismatched transplants from donors
to proteasomal degradation49. In the context of auto­ with genetically determined low HLA‑DPB1 expres­
immunity, N-terminal additions to a chromogranin A sion can lead to a high risk of GVHD if the recipients
peptide, an autoantigen in the NOD mouse model pre­ have high expression levels of HLA‑DPB1 (REF. 58).
sented on class II MHC molecules, could substantially Notably, genetic variants that reduce the expression of
improve T cell activation. This finding raises the pos­ HLA‑G, a nonclassical HLA molecule found on fetal
sibility that hybrid peptides presented by MHC class II extravillous trophoblast cells that invade the maternal
molecules could drive autoreactivity 50. A more recent decidua and generate the maternal–fetal interface, are
study in T1D has now shown that HLA class II proteins associated with pregnancy-related disorders such as
can indeed present spliced antigens that are generated pre-eclampsia59,60.
by covalent crosslinking of peptides51. Pathogenic CD4+
T cell clones isolated from NOD mice were found to HLA stability. Given the involvement of HLA expres­
recognize proinsulin hybrid peptides present in the sion levels in disease, post-translational regulation of
secretory granules of pancreatic β-cells. Furthermore, HLA stability has also been implicated in aberrant T cell
CD4+ T cells with an analogous specificity were also reactivity. The interdependence of HLA class I levels
isolated from the residual pancreatic islets of two organ and the peptide loading and binding process have long
donors with T1D. Therefore, T cell targeting of hybrid been recognized: the folding and assembly of class I
insulin peptides specifically present in the pancreas molecules in the endoplasmic reticulum (ER) lumen is
could provide an explanation for the breakdown of dependent on the multiprotein peptide-­loading com­
immune tolerance in T1D51 (TABLE 1). plex, and suboptimal assembly can promote ER reten­
tion or instability at the cell surface61. Tapasin, which
Regulation of HLA expression. In addition to the spe­ forms part of the peptide-loading complex, stabilizes the
cific presentation of antigens that trigger the stimulation peptide-free conformation of class I molecules, thereby
of autoreactive T cells, the level of expression of HLA increasing the chance of occupancy by a high-affinity
molecules at the cell surface, and thus the density of peptide62,63. Notably, HLA molecules that differ by only
relevant antigens, has been found to be associated with one amino acid residue in the peptide-­binding groove,
autoimmune diseases and other conditions (TABLE 1). For such as the HLA‑B*44:02 and HLA‑B*44:05 mole­
example, the distal intergenic XL9 element in the MHC cules, can differ strongly in their tapasin dependence,
locus has been found to alter the binding of interferon thus altering their trafficking to the cell surface63–65.
regulatory factor 4 (IRF4) and transcriptional regulator As HLA‑B*44:05 has a tyrosine rather than an aspar­
CTCF that modulates the transcription of HLA‑DRB1, tic acid at residue 116, it can constitutively acquire
HLA‑DQA1 and HLA‑DQB1, resulting in a 2.5‑fold peptides in a manner that is not dependent on the pep­
increase in the expression of HLA‑DR and HLA‑DQ tide-loading complex and is thus less susceptible to viral
proteins that predispose to systemic lupus erythema­ interference66.
tosus 52. Interestingly, genetically determined high Compared with HLA‑A and HLA‑B, HLA‑C proteins
expression levels of HLA‑C increase the risk of Crohn’s are present at the cell surface at a tenfold lower level67,
disease but also promote the control of infections such but these too can be differentially expressed depending
as HIV. Consequently, the opposing effects of differential on their peptide-binding capacity. HLA‑C*05 is found
HLA expression may exert a selection pressure in favour more abundantly at the cell surface than HLA‑C*07, as
of retaining allelic lineages with an HLA expression pro­ its peptide-binding cleft has a flatter structure, which
file that is associated with autoimmune disease but that is more permissive to the binding of a larger range of
protects against infection53. In a more extreme scenario, peptides that consequently help to stabilize the HLA
genetic defects that lead to partial or complete HLA molecule68 (FIG. 1i). HLA-derived peptides themselves can
class I or class II deficiency can cause bare lympho­cyte have an impact on HLA molecule stability. For example,
syndrome, which is typically characterized by extreme polymorphic HLA‑B leader peptide sequences can vary
Pancreatic islets
Clusters of different cell types susceptibility to bacterial, protozoan and viral infec­ in their capacity to bind HLA‑E and stabilize its expres­
found throughout the pancreas tions54. Moreover, many pathogen immune evasion strat­ sion levels at the cell surface, which in turn can skew NK
that include the egies include the downregulation of pathogen-­derived cell education towards that mediated by the binding of
insulin-producing β-cells, which peptide presentation by inhibiting HLA expression. HLA‑E to CD94/NKG2 receptors69.
are a main target of the
autoimmune response
For example, EBV can inhibit HLA class I synthesis Differential stability of HLA class II proteins has also
occurring in patients with through host shut-off 55 and encodes a repressor of MHC been reported and may have implications in disease
type 1 diabetes. class II transactivator (CIITA), thereby blocking HLA development. Antibody-dependent quantification of

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 7


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Allomorphs HLA‑DQ allomorphs on human primary B cells indicated Technological advances and challenges
Different MHC protein forms relatively little variation in the expression level of dif­ The identification of the numerous molecular mecha­
encoded by different alleles. ferent allomorphs70. However, a study using a cell-line- nisms by which HLA molecules are implicated in dis­
based MHC expression system suggests that HLA‑DQ ease development and resistance represents the outcome
allomorphs have a differential intrinsic stability. Some of several key technological advances in silico, in vitro
variation in the cell-surface density of HLA‑DQ mole­ (BOX 1) and in vivo (BOX 2) that are enabling key questions
cules may correlate with risk or resistance to auto­ regarding HLA biology to be addressed.
immune disorders 71. For example, T1D‑associated
HLA‑DQ proteins have been found to be unstable, Cross-comparing HLA genetic associations. Beyond
whereas T1D‑protective allomorphs are stable. Amino identifying HLA genetic associations with individual
acid variation at HLA‑DQ residue 47α, which is located diseases, the capacity to compare association patterns
on the outside of the peptide-binding groove, has been across autoimmune diseases can provide hints regard­
pinpointed as a key regulator of stability, potentially ing shared and distinct pathophysiological pathways
through a differential dependency on the MHC II pep­ that may have implications for understanding disease
tide-loading regulator HLA-DM72. In addition, evolu­ aetiology and therapy. In the future, such compari­
tionary analysis suggests that this resi­due was the target sons will be increasingly enabled by the availability
of positive diversifying selection71. HLA‑DQ instability of large resources, such as the UK Biobank (http://
may confer risk of autoimmune diseases by allowing www.uk­­biobank.ac.uk/), which allow for the simulta­
the escape of autoreactive T cells from thymic negative neous analysis of matched genetic and clinical infor­
selection but also by facilitating the formation of weak mation from different types of patients and controls77.
peptide–HLA complexes in the periphery with a wide Intriguing patterns include the observation that the
range of self peptides. The rela­tive instability of certain HLA class I risk alleles HLA‑B*27 (associated with
HLA class II proteins could explain why the same HLA ankylosing spondylitis), HLA‑C*06:02 (associated
alleles can be associated with either risk or resistance with psoriasis), HLA‑B*51 (associated with Behçet
to several different autoimmune diseases but cannot
explain all cross-disease association patterns. For exam­
Figure 2 | HLA–peptide–T cell receptor interactions can ▶
ple, although the HLA‑DRB1*15:01−HLA‑DQB1*06:02 promote autoimmunity. a | HLA class I allomorphs
haplotype, which encodes both the relatively less sta­ associated with autoimmune disease can promote the
ble HLA‑DRB1*15:01 protein and the intrinsically activation of autoreactive effector CD8+ T cells, the T cell
stable HLA‑DQB1*06:02 protein, is protective against receptors (TCRs) of which are generated stochastically in
T1D, autoimmune polyglandular syndrome and the thymus. Autoreactive HLA–peptide–TCR interactions
immuno­globulin A (IgA) deficiency 73–75, this haplotype may be affected by HLA gene expression levels, altered
confers risk of multiple sclerosis and narcolepsy 30,76. The peptide processing and the antigen affinity of
increased risk of multiple sclerosis can be associated autoreactive TCRs, ultimately leading to increased
with the less stable gene product of HLA‑DRB1*15:01; TCR-mediated activation of the autoreactive T cells. The
threshold for T cell activation may be further modulated
however, this is not the case for narcolepsy, in which
by non-HLA risk genes and environmental influences that
HLA‑DQ6‑mediated autoimmunity arises despite the collectively help lead to the development of a pathogenic
intrinsic high stability of the allomorph (TABLE 1). autoimmune response. b | HLA class II allomorphs
Collectively, these different mechanisms of HLA– associated with autoimmune disease can promote the
peptide–TCR binding may promote autoimmunity activation of autoreactive effector CD4+ T cells, the TCRs
by altering the capacity for autoreactive effector T cell of which are generated stochastically in the thymus.
activation and escape from the regulatory processes Autoreactive HLA–peptide–TCR interactions may be
that establish and maintain tolerance (FIG. 2). The sto­ affected by HLA gene expression levels and differential
chasticity of TCR generation probably contributes to the HLA protein stability, neoantigen creation (through
chance of an individual carrying autoreactive TCRs with protein post-translational modification and hybrid
peptide generation), ‘hotspot’ molecular mimicry,
an unconventional binding mode. However, auto­reactive
alternate TCR docking, TCR-mediated stabilization of
T cells exist not only in patients with auto­immune dis­ weak peptide–HLA complexes and an altered TCR binding
ease but also in healthy individuals with HLA risk allo­ register. The downstream consequence of these different
morphs; thus, the presence of autoreactive TCRs alone mechanisms is the increased TCR-mediated activation of
is unlikely to be sufficient for disease development. autoreactive effector T cells (and reduced generation or
Instead, this may further require numerous other genetic function of regulatory T cells in some instances). The
and environmental influences that modulate the avail­ threshold for T cell activation may be further modulated
ability, processing and presentation of relevant antigens by non-HLA risk genes and environmental influences that
as well as T cell activation thresholds. A fundamental collectively help lead to the development of a pathogenic
challenge for our understanding of how HLA–peptide– autoimmune response. α, MHC class II alpha chain;
β, MHC class II beta chain; β2m, β2-microglobulin;
TCR interactions promote disease is not only to deter­
APC, antigen-presenting cell; CLIP, class-II‑associated
mine how these different factors come together to break invariant chain peptide; ER, endoplasmic reticulum; ERAD,
self tolerance but also to elucidate the relative breadth ER-associated protein degradation; ERAP, ER
and importance of perturbed molecular mechanisms of aminopeptidase; MIIC, MHC-class-II‑containing
HLA–peptide–TCR binding in individual patients and compartment; miRNA, microRNA; TAP, transporter
across patient and control cohorts. associated with antigen processing.

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Unfolded protein response disease) and HLA‑A*29 (associated with birdshot homodimerization and expression levels at the cell
A cellular stress response chorio­retinopathy) all display epistatic interactions surface or an unfolded protein response79 (FIG. 2).
triggered because of the with ER aminopeptidase (ERAP) gene polymorphisms, For several of the conditions in which HLA class II
accumulation of unfolded or suggesting a key role for altered peptide processing 78. associations are the strongest determinants of disease
misfolded proteins within the
endoplasmic reticulum.
However, it remains to be elucidated whether these risk, interactions between different HLA class II alleles
interactions confer disease risk by leading to aber­ have been identified, as is the case for multiple sclero­
rant peptide presentation, changes in HLA class I sis30, rheumatoid arthritis, T1D and coeliac disease80.

a b
Nucleus
Transcription Altered HLA
expression

ER
Altered HLA gene
expression → impact Translation
of miRNAs +
HLA-DM
β α

Golgi

TAP Proteasome • Self proteins


• Viral proteins CLIP

HLA
class I
ERAP Early
ERAD MIIC endosome
Altered peptide
ER processing:
Golgi • Aberrant peptide Hybrid
presentation? peptides Protease
• Altered HLA
homodimerization? ‘Hotspot’
• Unfolded protein molecular Differential
response? mimicry HLA stability
HLA
APC class II APC
β2m
HLA class I

HLA–peptide–TCR Antigen
interaction TCR HLA–peptide– Post-translationally
TCR interaction modified protein
Low-affinity- CD3
mediated Microbial proteins
thymic escape containing cross-
reactive antigens
Low affinity for
LCK autoantigen LCK
enables thymic Alternate docking
escape
Higher affinity for
cross-reactive ↑ Cross-reactivity TCR stabilization ↑ Escape from
microbial peptide with microbial of weak peptide– thymic
peptide HLA complexes selection

Altered register
Differential generation
of autoreactive effector
versus regulatory T cells

Modulation by Increased TCR-mediated


non-HLA risk genes activation of autoreactive Modulation by
and environmental effector cells Increased TCR-mediated non-HLA genes
influences activation of autoreactive and environmental
effector cells influences

Autoimmunity Autoimmunity
CD8+ T cell CD4+ T cell

Nature Reviews | Immunology


NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 9
©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 1 | Studying HLA biology: the immunologist’s in silico and in vitro toolbox strategies86,87. In cancer, the study of HLA genetics has
not only focused on disease risk but has also found that
Genetics and genomics certain tumours display recurrent somatic mutations
Mapping the relationship of HLA alleles and haplotypes with single-nucleotide in HLA genes88, including mutations that promote a
polymorphisms (SNPs) in the MHC123 has revealed that, despite the high level of HLA loss of HLA allele heterozygosity 89; consequently, such
heterogeneity, the extensive linkage disequilibrium in the region allows for genetic
tumours are more likely to undergo immune eva­
variation to be captured through the use of tag SNPs, such that HLA alleles and
haplotypes can be imputed124. This has facilitated the interrogation of large case–control sion as there is less-efficient presentation of tumour
cohorts, leading to the accelerated discovery of HLA associations with even modest neoantigens. This has important thera­p eutic impli­
effect sizes across many different diseases. However, attributing associations to specific cations — particularly as a recent analysis has demon­
HLA alleles or haplotypes may not be as biologically informative as the capacity to strated that tumour HLA class  I genotype can be
pinpoint the exact polymorphisms implicated in disease risk. Identifying the precise used to predict which oncogenic mutations are more
coding or non-coding variants involved — by using improved analytical methods125 or likely to arise on the basis of the peptide sequences that
MHC deep sequencing126 — may provide further insight into peptide-binding groove are less effectively presented by the patient HLA class I
amino acid substitutions that could alter HLA–antigen interactions and into the molecules90.
differential regulation of HLA gene expression.
Tetramers Interrogating the antigen-specific T cell repertoire.
Peptide–HLA tetramers bind T cell receptors (TCRs) with a superior avidity compared Although HLA allele associations may suggest how
with peptide–HLA monomers (which exhibit a high rate of dissociation) and thus can specific HLA allomorphs contribute to disease suscep­
sensitively detect specific T cells present at a frequency as low as 0.02%. Moreover, even tibility and protection, more definitive evidence of their
lower frequency T cells can be detected when tetramer binding is coupled to magnetic
molecular mode of action requires the identification
enrichment techniques. The study of low-affinity peptide and HLA–peptide–TCR
interactions, such as those relevant in certain autoimmune diseases, still presents technical
of the rele­vant peptides and receptors that bind them.
problems127. This is particularly the case for HLA class-II‑restricted T cells, as the CD4 Exogenous and endogenous antigens presented by HLA
co-receptor does not facilitate the HLA–peptide–TCR interaction, leading to the molecules can vary according to tissue site, cellular route
underestimation of specific self-reactive T cell populations128. However, methods for of uptake and presentation, temporally owing to muta­
boosting HLA multimer staining are enabling the detection of T cells for which the TCR tion and modi­fication, and as an organism’s environ­
affinity for peptide–HLA complexes has a dissociation constant greater than 1 mM (REF. 94). mental exposures change, which all present technical
challenges. However, advances in proteomic and other
mass-spectrometry-based approaches are set to facili­
For coeliac disease, this may be due to HLA‑DQA1*05:01 tate the capturing of antigenic diversity, as evidenced by
and HLA‑DQB1*02:01 genotypes that are carried on dif­ the identification of novel hybrid peptides48. Crucially,
ferent haplotypes enabling the formation of HLA‑DQ2.5 these methods should be coupled to the investigation
heterodimers in trans81, whereas for multiple sclerosis, of the diseased tissue itself rather than peripheral blood
the protective effect of the HLA‑DQA1*01:01 haplotype alone, which is easily accessible but may be less inform­
in carriers of the disease-associated HLA‑DRB1*15:01 ative. There is a fundamental need to examine peptide
genotype30 may be related to TCR cross reactivity, as pro­ and TCR repertoires from the site of immunopathology,
posed for another multiple-sclerosis-related interaction where the disease-relevant epitopes and cells are local­
— HLA‑DRB1*15:01–HLA‑DRB5 (REF. 82). Intriguingly, ized. Moreover, even within a single individual, epitope
this latter HLA-DRB5 haplotype is also associated with spreading can either contribute to ongoing pathology or
Parkinson disease. Although Parkinson disease is con­ even have a protective effect91. In chronic conditions (for
sidered a primary neurodegenerative disorder, a recent example, coeliac disease), the range of epitopes driving
study has demonstrated that both HLA‑DRB1*15:01 pathological responses may change by epitope focus­
and HLA‑DRB5*01:01 can present peptides from ing, which is when T cell clones that are directed against
α‑synuclein, a protein aggregated in the brain of patients, immunodominant epitopes outcompete other clones92.
and the antigenic epitopes displayed can drive T cell Thus, antigenic diversity in the context of polymorphic
responses83. This suggests that the condition could have HLA molecules can promote the evolution of the T cell
an autoimmune component despite the absence of a repertoire, from the early events that occur in the thymus
prominent immune cell infiltrate. during the establishment of central tolerance through to
In the context of infectious disease, the coordi­ the events that occur over the course of a lifetime in the
nated study of host and viral genetic variation in dif­ periphery.
ferent ethnic groups is revealing the capacity of HLA The observations above indicate the value of moni­
micropolymorphisms to diversify the repertoire of toring and tracking specific T cells during the course
pathogen-specific cytotoxic lymphocytes, as exem­ of disease or upon vaccination or immunotherapy. The
Epitope spreading plified by infection with hepatitis B virus84. A recent development of HLA tetramers (BOX 1) and higher-­order
Describes how a self-directed
genome-wide association study of host and hepatitis multimers has been transformative for the study of T cell
immune response induced by a
single peptide (or epitope) C virus genomes indicated that different HLA alleles responses, as it has facilitated the ex vivo identification
could spread to include other also exert selective pressures on the hepatitis C virus and quantification of antigen-specific T cells as well,
peptides (or epitopes), not only genome, presumably via the impact of the adaptive as the mapping of their epitopes93,94. Ultimately, the
on the same autoantigen immune system, thereby driving in vivo viral evo­ combination of fluorescently labelled tetramer staining
(intramolecular spreading) but
also on other self molecules in
lution85. Thus, evaluating the relationship between with the interrogation of markers of cellular phenotype
close vicinity to the target cell HLA, T cell and virus genetic variation is important and function has yielded important insights into the
(intermolecular spreading). for designing effective vaccination and treatment dynamics of immune responsiveness in a wide range of

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

pathophysiological settings95, and the experimental use via distinct geometries that differ by a 15° rotation of
of peptide–HLA tetramers is increasingly being cou­ the whole TCR101. Other atypical MHC–peptide–TCR
pled to newer mass cytometry 96, TCR sequencing and interactions include reports of cross-class recognition
computational technologies97. (for example, when MHC class II molecules bind CD8+
TCRs)102 and the discovery of a reversed polarity of TCR
Exploiting the rules of HLA–peptide–T cell receptor recognition whereby TCR binding occurs with a 180°
engagement. Advances in deep-sequencing technolo­ rotation relative to the conventional binding mode103,104.
gies, particularly at the single-cell level98, are enabling the These examples challenge conventional views on the
detailed characterization of TCR sequence hetero­geneity, rules of TCR engagement; nevertheless, our growing
and this has spurred a concomitant effort to ascribe a understanding of the typical and atypical molecular
meaningful order to this variation. A recent study inte­ mechanisms of TCR recognition, in conjunction with
grating TCR sequences, TCR antigenic specificity (deter­ the application of next-generation sequencing methods,
mined by peptide–HLA tetramer panel screening) and may come to expedite the in silico identification of HLA–
structural data has shown that it is possible to group peptide–TCR binding partners and their correlation with
TCRs with shared specificities, both within and between cellular and clinical phenotypes through time.
individuals, on the basis of conserved motifs and CDR3
sequence similarity 97. The number and size of these Harnessing HLA function clinically
TCR groups may reflect immune response complexity, The progress in characterizing HLA diversity, HLA asso­
and clustering across individuals may suggest sharing of ciations with human disease and HLA–peptide–TCR
specif­icities for important antigens. This indicates that interactions and their mechanistic implications has
TCR sequence analysis may be of value clinically for galvanized one of the most fundamental goals of this
monitoring responses to infection or vaccination and area of research: to harness the improved understanding
that it might facilitate antigen discovery. of HLA function for clinical benefit (TABLE 2). With the
To investigate autoimmunity-prone T cell repertoires, current aim of achieving precision or even personalized
a study has explored the relative structural properties medicine, the capacity to stratify patients on the basis of
of TCRs and found that the presence of hydro­phobic their HLA genotypes and to administer antigen-specific,
residues at positions 6 and 7 of the β‑chain CDR3 loop patient-tailored disease prevention or immunotherapy
promotes reactivity to self antigens99. An investi­gation presents an especially attractive target.
of the specificity of peptide–MHC recognition by
TCRs in infection, through the use of TCR selection of Noncellular disease prevention strategies and thera­
peptide–MHC libraries in conjunction with deep pies: vaccination. Perhaps the most successful use of
sequencing, confirmed that for any given TCR inter­ antigen-­specific approaches for promoting human
action, there is a greater tolerance for variation of pep­ health is through vaccination to prevent the spread of
tide residues that are distal to the TCR interface. This infectious diseases. Despite substantial achievements,
was true to the extent that specific TCR ligands can be combating infections such as HIV, tuberculosis, hepa­
identified computationally 100. titis C and malaria is an ongoing challenge. However, for
Not all interactions between TCRs and peptide–HLA malaria, some progress has been reported based on the
molecules are confined to broad docking geometry con­ findings of a phase III trial of the RTS,S/AS01 candidate
straints. A notable example of a large-scale change in malaria vaccine105. Emerging infections, such as Ebola,
peptide–HLA binding geometry that is not easily pre­ Zika and Dengue viruses, highlight the need for rapid
dictable is that of the HLA‑A*02:01‑restricted melanoma vaccine generation and trialling strategies that rely on
DMF4 TCR; this TCR engages two homologous antigens the efficient identification of appropriate viral epitopes
to safely but effectively induce immunogenicity 106,107.
Even currently licensed vaccines have limitations
Box 2 | Studying HLA biology: the immunologist’s in vivo toolbox
that need to be overcome, such as the failure of recip­
Humanized mouse models ients to develop or maintain protective immunity and
The creation of humanized mouse models of disease, in which human genes or cells the occurrence of adverse events. For example, the per­
are transferred to mice to enable the functional dissection of specific HLA molecules, tussis resurgence observed in children and adolescents
has been instrumental to studying the biological consequences of disease-relevant who had previously received the acellular vaccine is at
antigen-specific responses in vivo129. The use of such models has facilitated the least partially related to waning immunity 108. For sev­
pinpointing of disease risk alleles that cannot be distinguished as pathophysiological
eral viral infections, including measles, mumps, rubella,
drivers by statistical means alone owing to high linkage disequilibrium. Interactions
between a multiple sclerosis HLA risk allele and disease-modifying alleles have been
influenza and hepatitis B virus, the induction and sus­
investigated in humanized mice and indicate the potential for antigenic-specific tainment of a humoral immune response following
immunomodulation82, as does the investigation of a Goodpasture-disease-protective vaccination correlates with specific HLA class I and II
HLA allele25. Studies in nonobese diabetic HLA‑A*02:01‑expressing mice have alleles that are determinants of nonresponsiveness or
identified CD8+ T cell populations that can also be found in patients with type 1 hyper-­responsiveness; this partly depends on the type
diabetes carrying HLA‑A*02:01, further suggesting that mechanistic insights relevant of T helper cells generated by specific HLA alleles109.
to human disease can be gained from the careful interrogation of humanized mice130. Antigenic changes in influenza virus require the
Developments in genetic engineering such as the use of CRISPR–Cas9 coupled to yearly reformulation and administration of vac­
improved microscopy techniques such as intravital imaging may further facilitate the cines. The 2009–2010 influenza A(H1N1)pdm09
study of antigen-specific immune responses in vivo.
(Pandemrix; GlaxoSmithKline) vaccination campaign

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 11


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 2 | Antigen-specific disease prevention and therapeutic strategies, desired mode of action and challenges
Preventative or therapeutic strategy Mode of action Development and implementation challenges
Noncellular
Vaccination against infection and cancer Boosting effector immune • Failure of development or maintenance of immune response
response • Emerging infections needing rapid antigen identification and trialling
• Unexpected side effects and differential efficacy
• Peptide and adjuvant selection
Peptide immunotherapy for allergy and Promoting peripheral • Peptide selection
autoimmune disease tolerance and/or pathogenic • Generation of desired type and magnitude of immune response
immune response dampening • Time of treatment relative to disease course
Peptide–HLA-coated nanoparticles for Promoting peripheral • Peptide selection
autoimmune disease tolerance • Generation of desired type and magnitude of immune response
• Time of treatment relative to disease course
• Trialling in humans required for safety and efficacy
Peptide–HLA-bearing exosomes for Boosting effector immune • Peptide selection
cancer response • Generation of desired type and magnitude of immune response
Engineered TCRs; for example, bispecific Promoting effector immune • Target selection and validation
ImmTACs with enhanced affinity and response • Temporal changes in target expression
T cell response redirection capacity for • Unwanted effects due to target expression on nonmalignant cells
cancer • Off-target effects
Cellular
DC adoptive transfer for cancer Boosting effector immune • Heterogeneity of DC molecular composition
immunotherapy response • Immunosuppression of DCs after transfer
• Poor storage capacity and handling difficulty
Regulatory T cell adoptive transfer for Promoting peripheral • Maintenance of functional stability
autoimmune disease and transplantation tolerance • Purity and storage
• Potency after transfer in pathophysiological milieu
T cell and/or total mononuclear Promoting effector immune • Tumour antigen loss
cell adoptive transfer for cancer response • Tumour immune evasion
immunotherapy • Peptide and adjuvant selection
• Immunosuppression of cells after transfer
DC, dendritic cell; ImmTACs, immune mobilizing monoclonal TCRs against cancer molecules; TCR, T cell receptor.

saw an unanticipated rise in the incidence of narcolepsy, thymic negative selection, the availability of peripheral
particularly in individuals carrying the predisposing neoantigens or inadequate Treg cell function.
HLA‑DQB1*06:02 allele110. Intriguingly, there is also Nevertheless, peptide immunotherapy with dis­
some evidence that Pandemrix administration inter­ ease-relevant autoantigenic epitopes has been con­
fered with the potential therapeutic benefit of the pep­ sidered as a potential option to treat autoimmune
tide immunotherapy GAD-alum (GAD formulated with disease112. A recent small placebo-controlled trial in
aluminium hydroxide; discussed below) in a phase III patients with newly diagnosed T1D has demonstrated
trial in individuals with new-onset T1D111. Collectively, that pro­insulin peptide immunotherapy is safe, does
these findings emphasize the importance of understand­ not expedite β-cell dysfunction and is correlated with
ing the factors that regulate vaccine immunogenicity, markers of immunomodulation 113. The efficacy of
including the recipient’s HLA genetic profile and prior antigen-­specific immunotherapy depends on adequate
immunogenic exposures. HLA-mediated antigen presentation to T cells. Boosting
of HLA–peptide–TCR interactions may be mediated
Noncellular disease prevention strategies and thera- by the use of adjuvants and small molecules that act as
pies: peptide immunotherapy. Despite the elucidation cell-surface HLA-loading enhancers, which have also
of many molecular mechanisms of HLA–peptide–TCR been tested in the context of cancer immunotherapy 114.
binding in the context of autoimmune disease, anti­ Notably, for GAD-alum administration to patients with
gen-specific immunotherapy for these conditions has T1D, there is some initial evidence that antigen presenta­
yet to become standard clinical practice. A key challenge tion may be further facilitated by direct intralymphatic
is determining whether peptide immunotherapy should injection of the therapeutic115.
aim either to delete effector T cells (to skew their patho­
genic phenotype towards a more anti-­inflammatory pro­ Noncellular disease prevention strategies and therapies:
file) or to promote Treg cell function (in order to prevent peptide–HLA delivery. A possible therapeutic strategy
disease progression due to continued organ damage). that bypasses the need for antigen uptake or cell-surface
This in turn depends on the nature of the HLA–peptide– loading that is associated with peptide immuno­therapies
TCR interactions that drive disease in any given indi­ is the use of peptide–HLA-coated nanoparticles. These
vidual and whether they are the outcome of insufficient can be tailored with respect to the peptide–HLA

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

molecules used and have been shown to drive the are not maintained; thus, the repertoire of neoantigen-­
expansion of antigen-specific regulatory responses in specific T cells must change through time in parallel
humanized mice116, although there are as yet no reports with newly occurring neoantigens121. This suggests the
of testing and efficacy in humans. need for strategies that broaden neoantigen-­specific
A similar approach is being trialled in malignancies T cell responses, especially as autologous patient T cells
including metastatic melanoma, advanced non-small- recognize only a fraction of predicted melanoma neo­
cell lung cancer and advanced colorectal cancer using antigens. To address this problem, a recent study
dendritic cell (DC)-derived exosomes, which are nano­ describes a system for identifying a larger number of
metre-sized membrane vesicles that express functional neoantigen-specific TCRs with the use of the naive
peptide–HLA complexes at their surface and which can T cell repertoire of healthy blood donors as a source of
induce T and NK cell-mediated immune responses in diverse TCRs restricted to the same HLA‑A molecule
patients117. Exosome-based immunotherapy is antigen-­ as the patient. The TCRs were then introduced into
specific and has distinct advantages over DC‑based can­ re­directed T cells by genetic engineering, and these cells
cer therapies, which are complicated by the difficulties ofwere capable of recognizing and killing patient-derived
culturing and storing DCs and high costs. melanoma cells expressing the relevant neoantigens122.
This approach therefore demonstrates the poten­
Noncellular disease prevention strategies and therapies: tial power of immunotherapeutic strategies in which
engineered T cell receptors. Other noncellular, antigen-­ HLA, antigen and TCR specificity are concomitantly
specific cancer immunotherapies include engineered considered and exploited for clinical benefit.
TCR-dependent strategies, such as the use of immune
mobilizing monoclonal TCRs against cancer (ImmTAC) Concluding remarks
molecules118. Native cancer-specific TCRs often have a After decades of studying HLA biology and its role in
weak affinity as a result of thymic selection, given that human disease, progress in genetic and molecular profil­
cancer-specific antigens are typically derived from self ing technologies, computational methods and structural
proteins; thus, boosting the activity of existing can­ analyses is broadening the scope of how the interactions
cer-specific T cells may be of limited benefit. However, between HLA molecules, the antigens they present and
ImmTACs are based on affinity-enhanced monoclonal the receptors that bind them can be utilized in a clini­
TCRs that bind their cognate peptide–HLA complexes cal setting. The many mechanisms identified to date by
with high, picomolar affinity. In addition to the specific which HLA–peptide–TCR interactions can promote
TCR-mediated binding of ImmTACs to tumour cells, disease represent individual case studies. Thus, it is still
these molecules are also fused to an anti‑CD3 anti­ unknown whether certain mechanisms are more com­
body fragment that then potentiates a redirected T cell monly utilized in particular diseases or in patient sub­
response and subsequent tumour cell killing 118. groups carrying the same HLA risk alleles or whether
multiple mechanisms may be implicated in individual
Cell-based therapies. DC‑based strategies to promote patients. Despite progress in identifying the specific
antigen-specific anticancer immune responses or toler­ HLA genes and alleles implicated in disease risk and
ogenesis are fraught with practical and economic chal­ resistance through large case–control cohort analyses, a
lenges, whereas T cell-based therapeutic approaches more high-throughput determination of the molecular
have shown more substantial promise. In the context of and cellular basis of these associations with respect to
autoimmune disease and transplantation, pilot studies the exact antigens and TCRs involved is a major goal
and early trials of the adoptive transfer of autologous, for the future. However, studying and monitoring HLA–
polyclonal Treg cells have provided support for the safety peptide–TCR interactions and their downstream impact
and possible capacity of these cells to induce tolerance in patient cohorts are becoming increasingly feasible
in vivo119,120. and will be required to meet the added challenges pre­
However, the greatest progress in the use of T cell- sented by the changing antigenic landscape within and
based therapies is in the cancer arena. An important across individuals. At the same time, ongoing research
finding has been the observation that successful treat­ efforts are already culminating in the use of antigen-­
ment of patients with melanoma with adoptive T cell specific disease prevention and therapeutic strategies,
therapy is driven by T cell reactivity against muta­ the design and implementation of which will probably
tion-derived neoantigens. As the neoantigens are lost be further aided by advances in genetic engineering and
owing to the impact of the T cells, the T cells themselves nanotechnology.

1. Amiel, J. in Study of the Leukocyte Phenotypes in developments in reporting HLA variation. 8. Sewell, A. K. Why must T cells be cross-reactive?
Hodgkin’s Disease in Histocompatibility Testing (ed. Hum. Immunol. 77, 233–237 (2016). Nat. Rev. Immunol. 12, 668–677 (2012).
Teraski, P. I.) 79–81 (Munksgaard, 1967). 5. Parham, P. & Ohta, T. Population biology of antigen 9. Quigley, M. F. et al. Convergent recombination shapes
2. Trowsdale, J. & Knight, J. C. Major presentation by MHC class I molecules. Science 272, the clonotypic landscape of the naive T‑cell repertoire.
histocompatibility complex genomics and human 67–74 (1996). Proc. Natl Acad. Sci. USA 107, 19414–19419
disease. Ann. Rev. Genom. Hum. Genet. 14, 6. Carrington, M. et al. HLA and HIV‑1: (2010).
301–323 (2013). heterozygote advantage and B*35‑Cw*04 10. Saunders, P. M. et al. A bird’s eye view of NK cell
3. Godfrey, D. I., Uldrich, A. P., McCluskey, J., disadvantage. Science 283, 1748–1752 receptor interactions with their MHC class I
Rossjohn, J. & Moody, D. B. The burgeoning family of (1999). ligands. Immunol. Rev. 267, 148–166
unconventional T cells. Nat. Immunol. 16, 1114–1123 7. Parham, P. & Moffett, A. Variable NK cell (2015).
(2015). receptors and their MHC class I ligands in 11. Hudson, L. E. & Allen, R. L. Leukocyte Ig‑Like
4. Robinson, J., Soormally, A. R., Hayhurst, J. D. & immunity, reproduction and human evolution. Receptors — a model for MHC class I disease
Marsh, S. G. E. The IPD-IMGT/HLA database — new Nat. Rev. Immunol. 13, 133–144 (2013). associations. Front. Immunol. 7, 281 (2016).

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 13


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

12. Fadda, L. et al. Peptide antagonism as a mechanism 36. Mohammed, F. et al. Phosphorylation-dependent 59. Ferreira, L. M. R., Meissner, T. B., Tilburgs, T. &
for NK cell activation. Proc. Natl Acad. Sci. USA 107, interaction between antigenic peptides and MHC Strominger, J. L. HLA‑G: at the interface of maternal-
10160–10165 (2010). class I: a molecular basis for the presentation of fetal tolerance. Trends Immunol. 38, 272–286
13. Sim, M. J. et al. Canonical and cross-reactive binding transformed self. Nat. Immunol. 9, 1236–1243 (2017).
of NK cell inhibitory receptors to HLA‑C allotypes is (2008). 60. Quach, K., Grover, S. A., Kenigsberg, S. &
dictated by peptides bound to HLA‑C. Front. Immunol. 37. Mannering, S. I. et al. The insulin A‑chain epitope Librach, C. L. A combination of single nucleotide
8, 193 (2017). recognized by human T cells is posttranslationally polymorphisms in the 3’ untranslated region of HLA‑G
14. Miles, J. J., McCluskey, J., Rossjohn, J. & Gras, S. modified. J. Exp. Med. 202, 1191–1197 (2005). is associated with preeclampsia. Hum. Immunol. 75,
Understanding the complexity and malleability of T‑cell 38. Molberg, O. et al. Tissue transglutaminase selectively 1163–1170 (2014).
recognition. Immunol. Cell Biol. 93, 433–441 (2015). modifies gliadin peptides that are recognized by gut- 61. Ljunggren, H. G. et al. Empty MHC class‑I molecules
15. Madsen, L. S. et al. A humanized model for multiple derived T cells in celiac disease. Nat. Med. 4, come out in the cold. Nature 346, 476–480
sclerosis using HLA‑DR2 and a human T‑cell receptor. 713–717 (1998). (1990).
Nat. Genet. 23, 343–347 (1999). 39. Bodd, M., Kim, C. Y., Lundin, K. E. & Sollid, L. M. T‑Cell 62. Wearsch, P. A. & Cresswell, P. Selective loading of
16. Hahn, M., Nicholson, M. J., Pyrdol, J. & response to gluten in patients with HLA‑DQ2.2 reveals high-affinity peptides onto major histocompatibility
Wucherpfennig, K. W. Unconventional topology of self requirement of peptide-MHC stability in celiac disease. complex class I molecules by the tapasin‑ERp57
peptide-major histocompatibility complex binding by a Gastroenterology 142, 552–561 (2012). heterodimer. Nat. Immunol. 8, 873–881 (2007).
human autoimmune T cell receptor. Nat. Immunol. 6, 40. Fallang, L. E. et al. Differences in the risk of celiac 63. Williams, A. P., Peh, C. A., Purcell, A. W., McCluskey, J.
490–496 (2005). disease associated with HLA‑DQ2.5 or HLA‑DQ2.2 & Elliott, T. Optimization of the MHC class I peptide
17. Bulek, A. M. et al. Structural basis for the killing of are related to sustained gluten antigen presentation. cargo is dependent on tapasin. Immunity 16, 509–
human beta cells by CD8+ T cells in type 1 diabetes. Nat. Immunol. 10, 1096–1101 (2009). 520 (2002).
Nat. Immunol. 13, 283–289 (2012). 41. Hovhannisyan, Z. et al. The role of HLA‑DQ8 β57 64. Rizvi, S. M. et al. Distinct assembly profiles of HLA‑B
18. Pugliese, A. et al. The insulin gene is transcribed in the polymorphism in the anti-gluten T‑cell response in molecules. J. Immunol. 192, 4967–4976 (2014).
human thymus and transcription levels correlate with coeliac disease. Nature 456, 534–538 (2008). 65. Thammavongsa, V., Raghuraman, G., Filzen, T. M.,
allelic variation at the INS VNTR‑IDDM2 susceptibility 42. Todd, J. A., Bell, J. I. & McDevitt, H. O. HLA‑DQ‑β Collins, K. L. & Raghavan, M. HLA‑B44
locus for type 1 diabetes. Nat. Genet. 15, 293–297 gene contributes to susceptibility and resistance to polymorphisms at position 116 of the heavy chain
(1997). insulin-dependent diabetes-mellitus. Nature 329, influence TAP complex minding via an effect on
19. Cole, D. K. et al. Hotspot autoimmune T cell receptor 599–604 (1987). peptide occupancy. J. Immunol. 177, 3150–3161
binding underlies pathogen and insulin peptide cross- 43. Corper, A. L. et al. A structural framework for (2006).
reactivity. J. Clin. Invest. 126, 2191–2204 (2016). deciphering the link between I-Ag7 and autoimmune 66. Zernich, D. et al. Natural HLA class I polymorphism
20. Yin, Y., Li, Y., Kerzic, M. C., Martin, R. & diabetes. Science 288, 505–511 (2000). controls the pathway of antigen presentation and
Mariuzza, R. A. Structure of a TCR with high affinity for 44. Broughton, S. E. et al. Biased T cell receptor usage susceptibility to viral evasion. J. Exp. Med. 200,
self-antigen reveals basis for escape from negative directed against human leukocyte antigen 13–24 (2004).
selection. EMBO J. 30, 1137–1148 (2011). DQ8‑restricted gliadin peptides is associated with 67. Apps, R. et al. Relative expression levels of the HLA
21. Quandt, J. A. et al. Unique clinical and pathological celiac disease. Immunity 37, 611–621 (2012). class‑I proteins in normal and HIV-infected cells.
features in HLA‑DRB1*0401‑restricted MBP 45. Petersen, J. et al. T‑Cell receptor recognition of J. Immunol. 194, 3594–3600 (2015).
111‑129‑specific humanized TCR transgenic mice. HLA‑DQ2‑gliadin complexes associated with celiac 68. Kaur, G. et al. Structural and regulatory diversity
J. Exp. Med. 200, 223–234 (2004). disease. Nat. Struct. Mol. Biol. 21, 480–488 shape HLA‑C protein expression levels. Nat. Commun.
22. Armstrong, K. M., Piepenbrink, K. H. & Baker, B. M. (2014). 8, 15924 (2017).
Conformational changes and flexibility in T‑cell 46. Dieterich, W. et al. Identification of tissue 69. Horowitz, A. et al. Class I HLA haplotypes form two
receptor recognition of peptide-MHC complexes. transglutaminase as the autoantigen of celiac disease. schools that educate NK cells in different ways. Sci.
Biochem. J. 415, 183–196 (2008). Nat. Med. 3, 797–801 (1997). Immunol. 1, eaag1672 (2016).
23. Stadinski, B. D. et al. Diabetogenic T cells recognize 47. Verpoort, K. N. et al. Isotype distribution of anti-cyclic 70. Duggal, P. et al. Genome-wide association study of
insulin bound to IAg7 in an unexpected, weakly binding citrullinated peptide antibodies in undifferentiated spontaneous resolution of hepatitis C virus infection:
register. Proc. Natl Acad. Sci. USA 107, 10978– arthritis and rheumatoid arthritis reflects an ongoing data from multiple cohorts. Ann. Intern. Med. 158,
10983 (2010). immune response. Arthritis Rheum. 54, 3799–3808 235–245 (2013).
24. Yang, J. et al. Autoreactive T cells specific for insulin (2006). 71. Miyadera, H., Ohashi, J., Lernmark, Å., Kitamura, T. &
B:11‑23 recognize a low-affinity peptide register in 48. Liepe, J. et al. A large fraction of HLA class I ligands Tokunaga, K. Cell-surface MHC density profiling
human subjects with autoimmune diabetes. Proc. Natl are proteasome-generated spliced peptides. reveals instability of autoimmunity-associated HLA.
Acad. Sci. USA 111, 14840–14845 (2014). Science 354, 354–358 (2016). J. Clin. Invest. 125, 275–291 (2015).
25. Ooi, J. D. et al. Dominant protection from HLA-linked This study demonstrates that the proteasome-­ 72. Zhou, Z. & Jensen, P. E. Structural characteristics of
autoimmunity by antigen-specific regulatory T cells. generated hybrid peptide pool accounts for as HLA‑DQ that may impact DM editing and
Nature 545, 243–247 (2017). much as one-third of the HLA class I susceptibility to type‑1 diabetes. Front. Immunol. 4,
This study reveals a mechanistic basis for the immunopeptidome, with implications for disease 262 (2013).
dominantly protective effect of HLA in autoimmune mechanisms and antigen-specific therapies. 73. Ferreira, R. C. et al. High-density SNP mapping of the
disease via an effect on self-epitope-specific 49. Hansen, T. H. & Bouvier, M. MHC class I antigen HLA region identifies multiple independent
Treg cells. presentation: learning from viral evasion strategies. susceptibility loci associated with selective IgA
26. Harkiolaki, M. et al. T cell-mediated autoimmune Nat. Rev. Immunol. 9, 503–513 (2009). deficiency. PLOS Genet. 8, e1002476 (2012).
disease due to low-affinity crossreactivity to common 50. Jin, N. et al. N‑Terminal additions to the WE14 74. Weinstock, C. et al. Autoimmune polyglandular
microbial peptides. Immunity 30, 348–357 (2009). peptide of chromogranin A create strong autoantigen syndrome type 2 shows the same HLA class II pattern
27. Sethi, D. K. et al. A highly tilted binding mode by a agonists in type 1 diabetes. Proc. Natl Acad. Sci. USA as type 1 diabetes. Tissue Antigens 77, 317–324
self-reactive T cell receptor results in altered 112, 13318–13323 (2015). (2011).
engagement of peptide and MHC. J. Exp. Med. 208, 51. Delong, T. et al. Pathogenic CD4 T cells in type 1 75. Hu, X. L. et al. Additive and interaction effects at three
91–102 (2011). diabetes recognize epitopes formed by peptide fusion. amino acid positions in HLA‑DQ and HLA‑DR
28. Wucherpfennig, K. W. & Strominger, J. L. Molecular Science 351, 711–714 (2016). molecules drive type 1 diabetes risk. Nat. Genet. 47,
mimicry in T cell-mediated autoimmunity: viral This study demonstrates that hybrid insulin 898–905 (2015).
peptides activate human T cell clones specific for peptides can be found in pancreatic islet β-cells in 76. Mignot, E. et al. Complex HLA‑DR and -DQ
myelin basic protein. Cell 80, 695–705 (1995). T1D and are antigenic for CD4+ T cells. interactions confer risk of narcolepsy-cataplexy in
29. Belbasis, L., Bellou, V., Evangelou, E., 52. Raj, P. et al. Regulatory polymorphisms modulate the three ethnic groups. Am. J. Hum. Genet. 68, 686–699
Ioannidis, J. P. A. & Tzoulaki, L. Environmental risk expression of HLA class II molecules and promote (2001).
factors and multiple sclerosis: an umbrella review of autoimmunity. eLife 5, e12089 (2016). 77. Cortes, A. et al. Bayesian analysis of genetic
systematic reviews and meta-analyses. Lancet Neurol. 53. O’Huigin, C. et al. The molecular origin and association across tree-structured routine healthcare
14, 263–273 (2015). consequences of escape from miRNA regulation by data in the UK Biobank. Nat. Genet. 49, 1311–1318
30. Moutsianas, L. et al. Class II HLA interactions HLA‑C alleles. Am. J. Hum. Genet. 89, 424–431 (2017).
modulate genetic risk for multiple sclerosis. Nat. (2011). 78. McGonagle, D., Aydin, S. Z., Gul, A., Mahr, A. &
Genet. 47, 1107–1113 (2015). 54. Shrestha, D., Szollosi, J. & Jenei, A. Bare lymphocyte Direskeneli, H. ‘MHC‑I‑opathy’-unified concept for
31. Nielsen, T. R. et al. Effects of infectious mononucleosis syndrome: an opportunity to discover our immune spondyloarthritis and Behcet disease. Nat. Rev.
and HLA‑DRB1*15 in multiple sclerosis. Mult. Scler. J. system. Immunol. Lett. 141, 147–157 (2012). Rheumatol. 11, 731–740 (2015).
15, 431–436 (2009). 55. Rowe, M. et al. Host shutoff during productive 79. Brown, M. A., Kenna, T. & Wordsworth, B. P. Genetics
32. Lang, H. L. E. et al. A functional and structural basis Epstein–Barr virus infection is mediated by BGLF5 of ankylosing spondylitis — insights into pathogenesis.
for TCR cross-reactivity in multiple sclerosis. Nat. and may contribute to immune evasion. Proc. Natl Nat. Rev. Rheumatol. 12, 81–91 (2016).
Immunol. 3, 940–943 (2002). Acad. Sci. USA 104, 3366–3371 (2007). 80. Lenz, T. L. et al. Widespread non-additive and
33. Babon, J. A. et al. Analysis of self-antigen specificity of 56. Balan, N., Osborn, K. & Sinclair, A. J. Repression of interaction effects within HLA loci modulate the risk of
islet-infiltrating T cells from human donors with type 1 CIITA by the Epstein–Barr virus transcription factor autoimmune diseases. Nat. Genet. 47, 1085–1090
diabetes. Nat. Med. 22, 1482–1487 (2016). Zta is independent of its dimerization and DNA (2015).
34. Koning, F., Thomas, R., Rossjohn, J. & Toes, R. E. binding. J. Gen. Virol. 97, 725–732 (2016). 81. Vader, W. et al. The HLA‑DQ2 gene dose effect in
Coeliac disease and rheumatoid arthritis: similar 57. Petersdorf, E. W. et al. HLA‑C expression levels define celiac disease is directly related to the magnitude and
mechanisms, different antigens. Nat. Rev. Rheumatol. permissible mismatches in hematopoietic cell breadth of gluten-specific T cell responses. Proc. Natl
11, 450–461 (2015). transplantation. Blood 124, 3996–4003 (2014). Acad. Sci. USA 100, 12390–12395 (2003).
35. Scally, S. W. et al. A molecular basis for the association 58. Petersdorf, E. W. et al. High HLA‑DP expression and 82. Gregersen, J. W. et al. Functional epistasis on a
of the HLA‑DRB1 locus, citrullination, and rheumatoid graft-versus-host disease. N. Engl. J. Med. 373, common MHC haplotype associated with multiple
arthritis. J. Exp. Med. 210, 2569–2582 (2013). 599–609 (2015). sclerosis. Nature 443, 574–577 (2006).

14 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

83. Sulzer, D. et al. T cells from patients with Parkinson’s 102. Hansen, S. G. et al. Cytomegalovirus vectors violate 121. Verdegaal, E. M. E. et al. Neoantigen landscape
disease recognize α-synuclein peptides. Nature 546, CD8+ T cell epitope recognition paradigms. dynamics during human melanoma‑T cell interactions.
656–661 (2017). Science 340, 1237874 (2013). Nature 536, 91–95 (2016).
This study demonstrates that the HLA associations 103. Beringer, D. X. et al. T cell receptor reversed 122. Stronen, E. et al. Targeting of cancer neoantigens with
with Parkinson disease may be related to the polarity recognition of a self-antigen major donor-derived T cell receptor repertoires. Science
presence of α‑synuclein-specific T cells, suggesting histocompatibility complex. Nat. Immunol. 16, 352, 1337–1341 (2016).
a relevance of the immune system in this 1153–1161 (2015). This study demonstrates that a higher frequency of
neurodegenerative condition. 104. Gras, S. et al. Reversed T cell receptor docking on a tumour mutations is more immunogenic than initially
84. Tan, A. T. et al. Host ethnicity and virus genotype major histocompatibility class I complex limits estimated and that healthy donors can be a valuable
shape the hepatitis B virus-specific T‑cell repertoire. involvement in the immune response. Immunity 45, source of T cells that are reactive to these tumour
J. Virol. 82, 10986–10997 (2008). 749–760 (2016). neoantigens, thereby indicating a new, personalized
85. Ansari, M. A. et al. Genome-to‑genome analysis 105. RTS,S Clinical Trials Partnership. Efficacy and safety of approach for cancer immunotherapy.
highlights the effect of the human innate and adaptive RTS,S/AS01 malaria vaccine with or without a booster 123. de Bakker, P. I. W. et al. A high-resolution HLA and
immune systems on the hepatitis C virus. Nat. Genet, dose in infants and children in Africa: final results of a SNP haplotype map for disease association studies in
49, 666–673 (2017). phase 3, individually randomised, controlled trial. the extended human MHC. Nat. Genet. 38,
86. Swadling, L. et al. A human vaccine strategy based on Lancet 386, 31–45 (2015). 1166–1172 (2006).
chimpanzee adenoviral and MVA vectors that primes, 106. Henao-Restrepo, A. M. et al. Efficacy and effectiveness 124. Dilthey, A., Cox, C., Iqbal, Z., Nelson, M. R. &
boosts, and sustains functional HCV-specific T cell of an rVSV-vectored vaccine expressing Ebola surface McVean, G. Improved genome inference in the MHC
memory. Sci. Transl. Med. 6, 261ra153 (2014). glycoprotein: interim results from the Guinea ring using a population reference graph. Nat. Genet. 47,
87. Park, S. H. et al. Subinfectious hepatitis C virus vaccination cluster-randomised trial. Lancet 386, 682–688 (2015).
exposures suppress T cell responses against 857–866 (2015). 125. Raychaudhuri, S. et al. Five amino acids in three HLA
subsequent acute infection. Nat. Med. 19, 107. Hadinegoro, S. R. et al. Efficacy and long-term safety proteins explain most of the association between MHC
1638–1642 (2013). of a Dengue vaccine in regions of endemic disease. and seropositive rheumatoid arthritis. Nat. Genet. 44,
88. Shukla, S. A. et al. Comprehensive analysis of cancer- N. Engl. J. Med. 373, 1195–1206 (2015). 291–296 (2012).
associated somatic mutations in class I HLA genes. 108. Plotkin, S. A. The pertussis problem. Clin. Infect. Dis. 126. Zhou, F. S. et al. Deep sequencing of the MHC region
Nat. Biotechnol. 33, 1152–1158 (2015). 58, 830–833 (2014). in the Chinese population contributes to studies of
89. McGranahan, N. et al. Allele-specific HLA loss and 109. Ovsyannikova, I. G., Dhiman, N., Jacobson, R. M. & complex disease. Nat. Genet. 48, 740–746 (2016).
immune escape in lung cancer evolution. Cell 171, Poland, G. A. Human leukocyte antigen 127. Dolton, G. et al. More tricks with tetramers: a practical
1259–1271.e11 (2017). polymorphisms: variable humoral immune responses guide to staining T cells with peptide-MHC multimers.
90. Marty, R. et al. MHC‑1 genotype restricts the to viral vaccines. Expert Rev. Vaccines 5, 33–43 Immunology 146, 11–22 (2015).
oncogenic mutational landscape. Cell 171, (2006). 128. Sabatino, J. J. Jr, Huang, J., Zhu, C. & Evavold, B. D.
1272–1283.e15 (2017). 110. Bomfim, I. L. et al. The immunogenetics of narcolepsy High prevalence of low affinity peptide-MHC II
91. Vanderlugt, C. L. & Miller, S. D. Epitope spreading in associated with A(H1N1)pdm09 vaccination tetramer-negative effectors during polyclonal CD4+
immune-mediated diseases: implications for (Pandemrix) supports a potent gene-environment T cell responses. J. Exp. Med. 208, 81–90 (2011).
immunotherapy. Nat. Rev. Immunol. 2, 85–95 (2002). interaction. Genes Immun. 18, 75–81 (2017). 129. Wekerle, H., Flugel, A., Fugger, L., Schett, G. &
92. Vader, W. et al. The gluten response in children with 111. Tavira, B. et al. Effect of simultaneous vaccination with Serreze, D. Autoimmunity’s next top models.
celiac disease is directed toward multiple gliadin and H1N1 and GAD-alum on GAD65‑induced immune Nat. Med. 18, 66–70 (2012).
glutenin peptides. Gastroenterology 122, response. Diabetologia 60, 1276–1283 (2017). 130. Yu, D. et al. Roquin represses autoimmunity by
1729–1737 (2002). 112. Larche, M. & Wraith, D. C. Peptide-based therapeutic limiting inducible T‑cell co‑stimulator messenger RNA.
93. Altman, J. D. et al. Phenotypic analysis of antigen- vaccines for allergic and autoimmune diseases. Nature 450, 299–303 (2007).
specific T lymphocytes. Science 274, 94–96 (1996). Nat. Med. 11, S69–S76 (2005).
94. Dolton, G. et al. Comparison of peptide-major 113. Alhadj Ali, M. et al. Metabolic and immune effects of Acknowledgements
histocompatibility complex tetramers and dextramers immunotherapy with proinsulin peptide in human new- C.A.D. is supported by the Wellcome Centre and the Royal
for the identification of antigen-specific T cells. Clin. onset type 1 diabetes. Sci. Transl. Med. 9, eaaf7779 Society. J.R. is supported by grants from the National Health
Exp. Immunol. 177, 47–63 (2014). (2017). and Medical Research Council (Australia), the Cancer Council
95. Nepom, G. T. MHC class II tetramers. J. Immunol. 114. Afridi, S., Hoessli, D. C. & Hameed, M. W. of Victoria, the Australian Research Council (ARC) and
188, 2477–2482 (2012). Mechanistic understanding and significance of small Worldwide Cancer Research and is an ARC Laureate Fellow.
96. Newell, E. W. et al. Combinatorial tetramer staining peptides interaction with MHC class II molecules for L.F. is supported by the Wellcome Centre, the Medical
and mass cytometry analysis facilitate T‑cell epitope therapeutic applications. Immunol. Rev. 272, Research Council, the Danish National Research Foundation,
mapping and characterization. Nat. Biotechnol. 31, 151–168 (2016). Takeda and the Oak Foundation.
623–629 (2013). 115. Ludvigsson, J., Wahlberg, J. & Casas, R.
97. Glanville, J. et al. Identifying specificity groups in the Intralymphatic injection of autoantigen in type 1 Author contributions
T cell receptor repertoire. Nature 547, 94–98 (2017). diabetes. N. Engl. J. Med. 376, 697–699 (2017). C.A.D. and J.R. researched data for the article; L.F., C.A.D.,
This study describes the development of an 116. Clemente-Casares, X. et al. Expanding antigen-specific J.R. and L.F. had a substantial input into the discussion of
algorithm for analysing large numbers of TCR regulatory networks to treat autoimmunity. Nature content; C.A.D., J.R and L.F. wrote the article; and L.F. and
sequences and defining TCR specificity groups 530, 434–440 (2016). C.A.D. carried out the review and editing.
across TCRs and individuals, which may facilitate This study demonstrates that systemic delivery of
analyses of T cell responses and ligand nanoparticles coated with autoimmune-disease-­ Competing interests statement
identification. relevant peptides bound to MHC class II molecules The authors declare no competing interests.
98. Stubbington, M. J. et al. T cell fate and clonality can trigger the generation and expansion of antige
inference from single-cell transcriptomes. n-specific regulatory CD4+ T cells, with potential Publisher’s note
Nat. Methods 13, 329–332 (2016). therapeutic implications. Springer Nature remains neutral with regard to jurisdictional
99. Stadinski, B. D. et al. Hydrophobic CDR3 residues 117. Pitt, J. M. et al. Dendritic cell-derived exosomes for claims in published maps and institutional affiliations.
promote the development of self-reactive T cells. cancer therapy. J. Clin. Invest. 126, 1224–1232
Nat. Immunol. 17, 946–955 (2016). (2016). Reviewer information
100. Birnbaum, M. E. et al. Deconstructing the peptide- 118. Liddy, N. et al. Monoclonal TCR-redirected tumor cell Nature Reviews Immunology thanks L. Klareskog, and two
MHC specificity of T cell recognition. Cell 157, killing. Nat. Med. 18, 980–987 (2012). anonymous reviewers, for their contribution to the peer
1073–1087 (2014). 119. Bluestone, J. A. et al. Type 1 diabetes immunotherapy review of this work.
101. Borbulevych, O. Y., Santhanagopolan, S. M., using polyclonal regulatory T cells. Sci. Transl. Med. 7,
Hossain, M. & Baker, B. M. TCRs used in cancer gene 315ra189 (2015). FURTHER INFORMATION
therapy cross-react with MART‑1/Melan‑A tumor 120. Chandran, S. et al. Polyclonal regulatory T cell therapy UK Biobank: http://www.ukbiobank.ac.uk/
antigens via distinct mechanisms. J. Immunol. 187, for control of inflammation in kidney transplants. Am.
ALL LINKS ARE ACTIVE IN THE ONLINE PDF
2453–2463 (2011). J. Transplant. 17, 2945–2954 (2017).

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 15


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like