You are on page 1of 7

MHC Class II Tetramers

Gerald T. Nepom

J Immunol 2012;188;2477-2482
This information is current as doi:10.4049/jimmunol.1102398
of March 23, 2012 http://www.jimmunol.org/content/188/6/2477

References This article cites 69 articles, 29 of which can be accessed free at:
http://www.jimmunol.org/content/188/6/2477.full.html#ref-list-1

Downloaded from www.jimmunol.org on March 23, 2012


Subscriptions Information about subscribing to The Journal of Immunology is online at
http://www.jimmunol.org/subscriptions
Permissions Submit copyright permission requests at
http://www.aai.org/ji/copyright.html
Email Alerts Receive free email-alerts when new articles cite this article. Sign up at
http://www.jimmunol.org/etoc/subscriptions.shtml/

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
9650 Rockville Pike, Bethesda, MD 20814-3994.
Copyright ©2012 by The American Association of
Immunologists, Inc. All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
MHC Class II Tetramers
Gerald T. Nepom
MHC class II tetramers have emerged as an important streptavidin molecules, forming ostensibly tetravalent com-
tool for characterization of the specificity and phenotype plexes; hence, the common use of the term “tetramers” for this
of CD4 T cell immune responses, useful in a large variety detection method.
of disease and vaccine studies. Issues of specific T cell Tetramer assays are widely used for single-cell phenotyping
frequency, biodistribution, and avidity, coupled with and enumeration and offer an important advantage over other
the large genetic diversity of potential class II restriction methods, such as ELISPOT and single-cell PCR, by enabling
elements, require targeted experimental design. Transla- the recovery and further study of sorted cells based on fluo-
tional opportunities for immune disease monitoring are rescent tetramer binding in flow cytometry. As a cytometry-
driving the rapid development of HLA class II tetramer based application, tetramers also provide the prospect of
ease of use and short assay time, analogous to Ab-based flow
use in clinical applications, together with innovations
cytometry studies. The major limitation to their use is the
in tetramer production and epitope discovery. The
requirement to have some knowledge of the specific pMHC

Downloaded from www.jimmunol.org on March 23, 2012


Journal of Immunology, 2012, 188: 2477–2482.
components involved in the recognition events being studied,
because these are necessary to construct the tetramer reagents.
A corollary to this element of MHC restriction is the con-

T
he concept of using soluble labeled ligands to detect cell
surface receptors is a standard approach for under- sideration that humans have very diverse HLA class II mole-
standing molecular specificity and function. However, cules, so that random sampling or clinical monitoring studies
only in the last 15 y has this general strategy been successfully may require the use of many different tetramers to include
applied to the analysis of Ag-specific T cells through the use a broad representation of the population.
of soluble MHC-peptide ligands that engage the abTCR. Straightforward solutions exist for both of these limita-
Originally developed for MHC class I recognition in the tions—HLA diversity and epitope identification—by making
context of CD8 T cells (1, 2), over the last decade similar the investment necessary to produce a universal set of class II
approaches have been successfully used for MHC class II tetramers for most HLA alleles, as well as by using functional
recognition in the context of CD4 T cells for a wide variety of selection of relevant epitopes as a criterion for study design.
Ags. From this experience, it is now evident that interrogating With these advances, discussed below, there has been a rapid
CD4 T cells using soluble tetramers has matured into a robust expansion of studies using tetramers to study CD4 T cell re-
technology, notably useful in numerous translational and sponse to vaccines, allergens, and a variety of Ags associated
clinical contexts. However, several constraints that limit their with autoimmune diseases. There has also been an improved
use are also now understood. In this Brief Review, key issues understanding surrounding the potential use of tetramers, be-
governing CD4 T cell identification by MHC class II tet- cause as the technological problems have been largely solved,
ramers are discussed, and examples from human immune there is now realization of some important biological barriers
monitoring studies illustrate the lessons learned. associated with interrogating the specific TCR–tetramer in-
teraction.
Adding specificity to the T cell flow cytometry toolkit
Peptide-MHC (pMHC) class II multimers display an anti- Rare CD4 T cell detection using class II pMHC tetramers
genic peptide in the class II-binding groove, functioning as A major challenge for studies using HLA class II tetramers is
a surrogate for recognition events that occur as part of the T the low frequency of Ag-specific CD4 T cells in peripheral
cell interaction with APCs. Various methods for displaying blood. Highly boosted responses, such as tetanus-specific or
pMHC class II molecules have been successfully used, ranging influenza-specific CD4 T cells after immunization, display
from monomeric pMHC bound to solid substrates or soluble transient levels of Ag-specific cells in the 1:1,000–1:20,000
in solution to higher-order multimers complexed with various range; specific memory T cells without recent boosting are
scaffold molecules. The most prevalent form of multimer in more often found in the 1:20,000–1:100,000 range; and rare
use today consists of biotin-labeled pMHC displayed on responses or autoantigen-specific responder cells are detectable

Benaroya Research Institute, Seattle, WA 98101 Abbreviations used in this article: pMHC, peptide-MHC; T1D, type 1 diabetes;
TGEM, tetramer-guided epitope mapping.
Received for publication December 7, 2011. Accepted for publication January 6, 2012.
Work in the author’s laboratory cited in this article was supported by grants from the Copyright Ó 2012 by The American Association of Immunologists, Inc. 0022-1767/12/$16.00
National Institutes of Health and the Juvenile Diabetes Research Foundation.
Address correspondence and reprint requests to Dr. Gerald T. Nepom, Benaroya Re-
search Institute, Immune Tolerance Network, 1201 Ninth Avenue, Seattle, WA 98101.
E-mail address: nepom@benaroyaresearch.org

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1102398
2478 BRIEF REVIEWS: MHC CLASS II TETRAMERS

in the 1:100,000–1:1 million range. Naive T cell frequencies not only are there multiple potential protein targets within
to particular pMHC are also ∼1:200,000–1:1 million. Be- each pathogen, but a huge number of processed and presented
cause of these low cell frequencies, the first generation of peptides are potentially targets for CD4 T cell recognition.
tetramer-based immunoassays used in vitro activation and/or For example, within the protective Ag of Bacillus anthracis,
expansion to enable T cell detection (3), somewhat analo- multiple epitopes are present; as expected, the dominance of
gous to the historically used techniques of limiting dilution- each epitope varies depending on the HLA genotype of the
proliferation assays and ELISPOT analysis. Of course, a ma- individual subject (23). Therefore, selection of tetramers for
jor problem with such an approach was the potential for studies of the T cell response to anthrax or similarly complex
phenotypic changes introduced by in vitro manipulation; agents involves matching the HLA class II genotype of each
therefore, these studies were limited primarily to the estimates subject with appropriately tailored pMHC tetramers (31). In
of cell frequency and specificity. contrast, occasionally there are immunodominant epitopes
Current second-generation tetramer assays avoid the use of from pathogens that bind promiscuously to multiple HLA
in vitro expansion and are either based on single-cell capture class II molecules. When this occurs, production and use of
technology (4–6) or on direct flow cytometry staining analysis appropriate pMHC tetramers becomes straightforward. For
(7–10). In the latter, stringent “dumping” criteria and en- example, the hemagglutinin protein of influenza A H1N1
richment with magnetic bead-trapping procedures are used to contains a peptide epitope (aa 306–319) that binds to at least
remove cells that lack tetramer specificity, enriching the Ag- five common human class II molecules and is recognized by
specific population as much as 10,000-fold, allowing for de- T cells in each case. Most people, through either natural ex-
tection of rare events (e.g., 10 tetramer-binding T cells from posure or periodic flu vaccination, contain peripheral CD4
20 ml of peripheral blood). In addition, multiparameter flow T cells recognizing this epitope (14). A similar strategy has been

Downloaded from www.jimmunol.org on March 23, 2012


cytometry analysis using a variety of mAbs simultaneously adopted to search for conserved epitopes in pandemic strain
with tetramer assays allows for direct phenotyping of the Ag- H5N1, after vaccination (32). T cells recognizing additional
specific cells, a major advance toward discriminating T cell influenza epitopes can be found simultaneously in the same
functional subsets directed to a single Ag. blood samples using other tetramers but appear to be more
HLA class II tetramers are now widely used in studies of specific to a given individual (12, 33). An interesting excep-
pathogen immunity and vaccine development, in evaluation of tion was seen in tetramer analysis with pMHC corresponding
antitumor responses, in allergy monitoring and desensitization to the H5N1 influenza strain hemagglutinin, with cross-re-
studies, and in autoimmunity (Fig. 1). Examples of each are activity from pre-existing immunity to other influenza strains,
summarized below and illustrate particular issues faced in allowing detection of T cells reactive to H5N1, even in the
these different applications. absence of direct exposure to this strain (24).
Monitoring immunity to infectious pathogens. Class II tetramers With hundreds of potential peptide epitopes to choose
have been used for analysis of a variety of human CD4 T cell from, selecting pMHC tetramers for studying infectious patho-
responses to pathogens, including influenza A, Borrelia, EBV, gens or vaccines is challenging. Traditional methods involving
CMV, Mycobacterium tuberculosis, human T-lymphotropic functional testing of individual peptides from large libraries
virus 1, hepatitis C, anthrax, severe acute respiratory syn- are laborious and, when based on a particular functional param-
drome virus, human papillomavirus, and HIV (3, 7, 11–30). eter (e.g., IFN ELISPOT analysis), may omit epitopes that are
Ag-specific T cells are detected in peripheral blood, with a very important for other T cell subsets or lineages. To address this
broad range of frequency, depending on prior Ag exposure, issue, an unbiased platform has been developed for a tetramer-
vaccination history, and timing of immunization. The most binding and flow cytometry screening method that directly
significant limitation for these studies is simply the large determines suitable pMHC tetramers. This technique, called
number of potential epitopes present in complex organisms; tetramer-guided epitope mapping (TGEM), involves the dis-
play of large peptide libraries loaded into HLA class II mol-
ecules (Fig. 2). Pools of tetramers are used to probe the T cell
population of interest, and positive tetramer binding, assessed
by flow cytometry, is deconvoluted in a second flow cytometry
step into individual pMHC targets (34, 35). This approach
has been applied to a variety of infectious agents, with epitope
identification information deposited in the National Institute
of Allergy and Infectious Diseases-sponsored Immune Epitope
Database and Analysis Resource, IEDB.org.
Allergen tetramers: a tool for improved immunotherapy? Transla-
tion from a laboratory tool to a clinically useful biomarker is
a difficult transition for any technology, but in the area of
allergy desensitization therapy, human class II tetramers have
their most immediate translational potential. Similar to the
study of infectious pathogens, tetramer development for
allergens is complicated by the diversity and number of po-
FIGURE 1. Molecular design schematic of a class II tetramer molecule,
illustrating four pMHC domains tethered to a central streptavidin molecule
tential CD4 T cell epitopes. However, for many of the major
through flexible linker sequences and leucine zipper motifs to provide allergic disorders, including allergies to foods, pollen, and
structural stability. Some of the major areas of tetramer analysis in human dander, there is a large body of knowledge narrowing the
disease applications are listed. field of target allergen proteins. The TGEM technique has
The Journal of Immunology 2479

FIGURE 2. Epitope identification using a tetramer-


guided mapping strategy. Large peptide libraries, sep-
arated into sets of pooled sequences, are loaded into the
binding groove of recombinant class II molecules and
assembled into tetramers. Direct flow cytometry analysis
of T cell populations identifies peptides containing
actual epitopes representative of the natural immune
response.

been applied to several of these, resulting in the identification sulin, IA-2, and ZnT8 have all been used to evaluate T cell

Downloaded from www.jimmunol.org on March 23, 2012


of epitopes and tetramers suitable for analysis of peripheral frequencies in peripheral blood. Findings from these studies
T cell recognition to major allergens (10, 16, 26, 29). In support the concept that the autoreactive repertoire matures
allergic individuals, the frequency of tetramer-binding CD4 during disease progression. First, normal individuals, as well as
T cells in peripheral blood, particularly during periods of T1D subjects, have low, but detectable, levels of circulating
allergen exposure, is suitably high, allowing direct detection tetramer-binding CD4 cells. However, these display markers
by flow cytometry after tetramer binding. In a recent study of naive lymphocytes in the former group, whereas they are
by Wambre et al. (10), multiparameter flow analysis docu- predominantly memory phenotype in the latter, presumably
mented progressive shifts in cell surface marker profiles reflect a history of prior activation in vivo. Second, although
associated with T cell deviation during s.c. allergen immu- some high-avidity autoreactive T cells are present and can be
notherapy. Current clinical desensitization protocols use expanded in culture, the population of tetramer-binding cells
allergen extracts, require very long-term administration, and contains both low- and high-avidity TCR for the autoantigen
carry significant risk for adverse allergic response, making it pMHC. By taking advantage of the unique genetics of hu-
attractive to consider using tetramer-based immune monitor- man T1D, in which a polymorphism in the proinsulin pro-
ing in this context. Also, because one approach to improve moter region controls levels of thymic expression (48, 49),
allergy immunotherapy is to use peptides instead of allergen Durinovic-Belló et al. (44) demonstrated that the strength of
extracts (19, 36), the TGEM technique and tetramer mon- tetramer binding to proinsulin-specific T cells correlated with
itoring may help to identify appropriate peptide epitopes for this genotype, directly linking thymic Ag expression with
use, particularly if stratification based on HLA genotype is avidity selection for the CD4 repertoire. Third, when T1D
needed. patients undergo pancreas transplantation, they essentially
Tetramer profiling of low-avidity T cells in autoimmunity. Human receive an immunological challenge with islet autoantigens, in
class II pMHC tetramers containing autoantigenic specificities addition to the alloreactivity associated with an organ trans-
have been used to study T cell responses in a variety of dis- plant. Tetramer analysis of such subjects, followed for several
eases, including type 1 diabetes (T1D), celiac disease, pemphi- years after transplantation, documented both the expansion
gus vulgaris, rheumatoid arthritis, multiple sclerosis, and uve- of autoantigen pMHC tetramer-binding CD4 T cells and,
itis (37–47). These studies have highlighted a fundamental more remarkably, the persistence of unique clonotypic TCR
point: the avidity of the TCR interaction with its pMHC sequences within this population, indicating a recurrence of
target is influenced by selection against high-affinity self- memory responses from a pre-existing autoreactive repertoire
Ag recognition. Unlike tetramer analyses in the infectious (50, 51).
pathogen and allergen studies cited above, in which most of The low frequency and low avidity of T cells specific
the responding T cells have a high-avidity binding to ap- for autoantigens present special technical challenges, but sev-
propriate pMHC tetramers, tetramer binding to peripheral eral creative approaches have shown promise for improving
T cells in the context of autoimmunity displays a much wider tetramer-detection strategies. In a recent study of subjects
spectrum of relative strength of interaction. In addition, the with rheumatoid arthritis, class II tetramers were used to test
frequency of autoreactive CD4 T cells specific for a particular an intriguing mechanistic hypothesis: posttranslational mod-
pMHC–self-Ag complex is quite low in peripheral blood, ification of peptide epitopes creates neoantigenic specific-
often less than five per million lymphocytes, requiring large ities targeted in the disease setting (52). James et al. (45)
sample volumes and careful handling for reliable detection. also prepared HLA class II tetramers containing citrulline-
The most complete autoimmunity studies have been per- modified peptides based on fibrinogen protein sequences
formed in the context of T1D, a disease in which several major found in joint tissue, and they documented the presence of
autoantigens are well characterized. Class II tetramers specific T cells that bound these tetramers and responded to the
for the T1D autoantigens glutamate dehydrogenase, proin- corresponding autoantigen. In celiac disease, where there is
2480 BRIEF REVIEWS: MHC CLASS II TETRAMERS

also posttranslational modification of target Ags, class II tet- quence or a too-stringent binding register is a potential con-
ramers composed of gliadin pMHC complexes were shown to cern. An important use of noncovalent peptide-loaded
bind peripheral CD4 T cells; however, these cells were only pMHC production systems is the ability to assess many dif-
detectable when the subjects consumed gluten-containing ferent tetramer-binding specificities at the same time. This has
bread for 3 d (53). This finding strongly suggests that mo- been exploited in the TGEM technique described above, in
bilization of the Ag-specific cells from tissue to blood was which large mixtures of peptides, most commonly repre-
necessary for visualization, and it may provide a generalizable senting the entire protein sequence of potential antigenic
strategy, involving Ag challenge, for improving the detection targets, are interrogated to determine which peptides within
threshold for rare autoreactive cells. the mixture contain actual T cell epitopes.
A related issue, particularly relevant to the analysis of low- Because of the high specificity and sensitivity of TCR rec-
avidity T cell responses in autoimmunity, is the definition of ognition for pMHC, tetramer-binding studies can also be
a precise peptide-binding register for docking within the class designed to identify unconventional antigenic epitopes. For
II molecule. The presence of more than one binding register example, many protein therapeutics that are administered to
within long peptides generated during Ag processing can result patients are potentially immunogenic. Analysis of the T cell
in presentation of pMHC ligands to distinct TCR, with po- response in these cases, using tetramers containing pMHC
tentially different roles in selection and autoreactivity (54, 55). specificities from the therapeutic molecule, can readily identify
Tetramer studies of tumor Ags are similar to the autoantigen the specific epitopes that trigger immunogenicity, as illustrated
approach, with an opportunity to help inform both disease in a study of Factor VIII administration in hemophilia A (63).
mechanism and therapy. Although most tumor Ags are self-Ags Examples of technical variations that are designed to improve
or modified self-Ags, intentional immunization with these Ags the efficiency of tetramer production include coexpression

Downloaded from www.jimmunol.org on March 23, 2012


is conducted for therapeutic benefit. For example, class II with chaperone proteins (64), biotinylation of the expressed
tetramer studies in patients with malignant melanoma after MHC molecule during synthesis (65), modifications of peptide
experimental vaccine therapies documented evidence of im- linkers used to dock covalent peptides in the coexpressed MHC
munogenicity, measured the effects of adjuvant on T cell groove (66), or the use of cleavable CLIP peptides (7), which
lineage, and have been used to evaluate shifts in T cell sub- take advantage of the natural role of invariant chain residues
sets (56–59). Indeed, because the pMHC complex is itself (CLIP) that are a functional intermediate in loading peptides
a surrogate for Ag recognition, it is feasible that tetramers into class II molecules. One of the most intriguing innovations
might be considered for therapeutic expansion of Ag-specific is the use of entire libraries of class II pMHC molecules plated
effector T cells (60). on arrays designed to capture Ag-specific T cells (67), similar to
earlier studies using class I tetramers (68).
Variations on the tetramer theme
Biophysical studies and off-rate analysis suggest that multi-
valent binding of pMHC to the TCR is essential for successful Conclusions
tetramer imaging of Ag-specific T cells. Monomeric and di- After more than a decade of experience, class II tetramer
meric forms of pMHC are much less efficient than tetramers analysis has become a regular component of T cell-specificity
(61), whereas higher-order multimers, such as octomers, studies across the entire landscape of immunological investi-
decamers, or pMHC-coated beads, are all successful reagents, gation. However, a decision to include class II tetramer studies,
generally comparable to tetramers. However, a caveat to this as well as how to use them, is not as routine as selecting Abs for
interpretation is that streptavidin aggregates during storage, so flow cytometry. There are four key issues that should be
that experiments ostensibly using tetrameric forms of pMHC considered and used as the basis for experimental choice.
linked to streptavidin may, in fact, be using higher-order First, what is the anticipated frequency of CD4 T cells
complexes, at least in part. recognizing immunodominant epitopes in the system? If
A variety of methods has been successfully used to manu- there is no prior evidence for immunodominance and no
facture pMHC class II tetramers, with expression of HLA information from previous T cell assays regarding prevalence,
class II molecules as “empty” (without intentional loading then it is a challenge to select a particular pMHC tetramer that
of peptide into the class II Ag-binding groove); “loaded,” will be representative of the T cell response profile. Either
in which the expression system is coupled to a facilitated multiple different pMHC can be selected, individually or as
peptide-loading process, resulting in production of complete a mixed tetramer pool, or a technique such as TGEM can be
pMHC; or “covalent,” in which an MHC expression vector used in pilot studies to directly determine the appropriate
is engineered to cotranslationally express the peptide, with pMHC combinations.
a flexible and/or removable linker sequence allowing for 1:1 Second, what is the likely biodistribution of CD4 T cells of
stoichiometry of peptide/MHC and directed peptide bind- interest? In cases such as celiac disease, as described above,
ing into the class II-binding groove. Expression systems have mobilization of the Ag-specific T cells into the bloodstream
included mammalian, insect cell, or bacterial sources (62). was accomplished by Ag challenge. This may have general
There are advantages to each system, depending on the in- applicability, supported by the observations in T1D that
tended type of tetramer assay planned. For example, covalent tetramer-positive cells are much higher after pancreas trans-
pMHC are well suited for consistent production systems and plantation or after glutamate dehydrogenase immunization
for situations in which a small number of specific pMHC (H. Reijonen, personal communication) compared with un-
combinations are used. They are not well suited to screening immunized diabetic subjects. If the Ag-specific cells are at tis-
assays involving hundreds of different potential peptide Ags, sue sites of interest, more complex biopsy and in situ histo-
and neoepitope exposure due to the presence of linker se- chemistry techniques are needed.
The Journal of Immunology 2481

Third, avidity matters. Unlike foreign Ag responses in vac- specific CD4(+) T-cell outcomes during specific immunotherapy. J. Allergy Clin.
Immunol. 129: 544–551.
cine or allergy studies, self-Ag responses in autoimmunity or 11. Meyer, A. L., C. Trollmo, F. Crawford, P. Marrack, A. C. Steere, B. T. Huber,
tumor immunology reflect a diverse spectrum of TCR avidity J. Kappler, and D. A. Hafler. 2000. Direct enumeration of Borrelia-reactive CD4
for pMHC complexes, including interactions that are near or T cells ex vivo by using MHC class II tetramers. Proc. Natl. Acad. Sci. USA 97:
11433–11438.
below the threshold for robust tetramer-binding detection. If 12. Danke, N. A., and W. W. Kwok. 2003. HLA class II-restricted CD4+ T cell
the experimental system requires detection of such low-avidity responses directed against influenza viral antigens postinfluenza vaccination. J.
Immunol. 171: 3163–3169.
interactions, methods to amplify the tetramer-binding signals 13. Lucas, M., C. L. Day, J. R. Wyer, S. L. Cunliffe, A. Loughry, A. J. McMichael, and
may be helpful. Alternatively, T cell activation triggered by P. Klenerman. 2004. Ex vivo phenotype and frequency of influenza virus-specific
tetramer binding can be used as a surrogate measure, with a CD4 memory T cells. J. Virol. 78: 7284–7287.
14. Ye, M., S. Kasey, S. Khurana, N. T. Nguyen, S. Schubert, C. T. Nugent, K. Kuus-
threshold lower than conventional flow cytometry detection Reichel, and J. Hampl. 2004. MHC class II tetramers containing influenza hem-
(62, 69). agglutinin and EBV EBNA1 epitopes detect reliably specific CD4(+) T cells in
healthy volunteers. Hum. Immunol. 65: 507–513.
Fourth, how genetically diverse is the population being 15. Bronke, C., N. M. Palmer, G. H. Westerlaken, M. Toebes, G. M. van Schijndel,
studied? The requirement for pMHC cognate recognition by the V. Purwaha, K. E. van Meijgaarden, T. N. Schumacher, D. van Baarle, K. Tesselaar,
CD4 T cell dictates that at least one of the class II molecules in and A. Geluk. 2005. Direct ex vivo detection of HLA-DR3-restricted cytomega-
lovirus- and Mycobacterium tuberculosis-specific CD4+ T cells. Hum. Immunol. 66:
each subject corresponds to the HLA molecule used for the 950–961.
tetramer. For populations that are highly enriched for particular 16. Macaubas, C., J. Wahlstrom, A. P. Galvão da Silva, T. G. Forsthuber,
G. Sønderstrup, W. W. Kwok, R. H. DeKruyff, and D. T. Umetsu. 2006. Allergen-
HLA class II genes, such as in T1D, rheumatoid arthritis, multiple specific MHC class II tetramer+ cells are detectable in allergic, but not in non-
sclerosis, or celiac disease, this is not a limiting factor. However, allergic, individuals. J. Immunol. 176: 5069–5077.
for population-based vaccine studies or allergy monitoring trials, 17. Ulsenheimer, A., M. Lucas, N. P. Seth, J. Tilman Gerlach, N. H. Gruener,
A. Loughry, G. R. Pape, K. W. Wucherpfennig, H. M. Diepolder, and
a large number of tetramers is needed or, alternatively, subjects P. Klenerman. 2006. Transient immunological control during acute hepatitis C

Downloaded from www.jimmunol.org on March 23, 2012


are preselected based on HLA genotype. Either way, a signifi- virus infection: ex vivo analysis of helper T-cell responses. J. Viral Hepat. 13: 708–
714.
cant complexity is added to the study. HLA class II tetramers 18. Höhn, H., C. Kortsik, I. Zehbe, W. E. Hitzler, K. Kayser, K. Freitag, C. Neukirch,
for 25 of the most common genotypes are now available, so this P. Andersen, T. M. Doherty, and M. Maeurer. 2007. MHC class II tetramer guided
is not an insurmountable barrier, if taken into account in experi- detection of Mycobacterium tuberculosis-specific CD4+ T cells in peripheral blood
from patients with pulmonary tuberculosis. Scand. J. Immunol. 65: 467–478.
mental design (http://www.benaroyaresearch.org/our-research/ 19. Kinnunen, T., K. Jutila, W. W. Kwok, M. Rytkönen-Nissinen, A. Immonen,
core-resources/tetramer-core-laboratory). S. Saarelainen, A. Närvänen, A. Taivainen, and T. Virtanen. 2007. Potential of an
altered peptide ligand of lipocalin allergen Bos d 2 for peptide immunotherapy. J.
Allergy Clin. Immunol. 119: 965–972.
Acknowledgments 20. Laughlin, E. M., J. D. Miller, E. James, D. Fillos, C. C. Ibegbu, R. S. Mittler,
The work of numerous colleagues at the Benaroya Research Institute and else- R. Akondy, W. Kwok, R. Ahmed, and G. Nepom. 2007. Antigen-specific CD4+
where, who have pioneered the class II tetramer technology, is gratefully ac- T cells recognize epitopes of protective antigen following vaccination with an an-
knowledged, and any omissions from the citation list, limited by space, are thrax vaccine. Infect. Immun. 75: 1852–1860.
21. Nose, H., R. Kubota, N. P. Seth, P. K. Goon, Y. Tanaka, S. Izumo, K. Usuku,
inadvertent. Y. Ohara, K. W. Wucherpfennig, C. R. Bangham, et al. 2007. Ex vivo analysis of
human T lymphotropic virus type 1-specific CD4+ cells by use of a major histo-
compatibility complex class II tetramer composed of a neurological disease-
Disclosures susceptibility allele and its immunodominant peptide. J. Infect. Dis. 196: 1761–
The author has no financial conflicts of interest. 1772.
22. Ebinuma, H., N. Nakamoto, Y. Li, D. A. Price, E. Gostick, B. L. Levine, J. Tobias,
W. W. Kwok, and K. M. Chang. 2008. Identification and in vitro expansion of
References functional antigen-specific CD25+ FoxP3+ regulatory T cells in hepatitis C virus
infection. J. Virol. 82: 5043–5053.
1. Altman, J. D., P. A. H. Moss, P. J. R. Goulder, D. H. Barouch, M. G. McHeyzer-
23. Kwok, W. W., J. Yang, E. James, J. Bui, L. Huston, A. R. Wiesen, and M. Roti.
Williams, J. I. Bell, A. J. McMichael, and M. M. Davis. 1996. Phenotypic analysis
2008. The anthrax vaccine adsorbed vaccine generates protective antigen (PA)-
of antigen-specific T lymphocytes. Science 274: 94–96.
2. Lebowitz, M. S., S. M. O’Herrin, A. R. Hamad, T. Fahmy, D. Marguet, Specific CD4+ T cells with a phenotype distinct from that of naive PA T cells.
N. C. Barnes, D. Pardoll, J. G. Bieler, and J. P. Schneck. 1999. Soluble, high- Infect. Immun. 76: 4538–4545.
affinity dimers of T-cell receptors and class II major histocompatibility complexes: 24. Roti, M., J. Yang, D. Berger, L. Huston, E. A. James, and W. W. Kwok. 2008.
biochemical probes for analysis and modulation of immune responses. Cell. Healthy human subjects have CD4+ T cells directed against H5N1 influenza virus.
Immunol. 192: 175–184. J. Immunol. 180: 1758–1768.
3. Novak, E. J., A. W. Liu, G. T. Nepom, and W. W. Kwok. 1999. MHC class II 25. Yang, J., E. James, M. Roti, L. Huston, J. A. Gebe, and W. W. Kwok. 2009.
tetramers identify peptide-specific human CD4(+) T cells proliferating in response Searching immunodominant epitopes prior to epidemic: HLA class II-restricted
to influenza A antigen. J. Clin. Invest. 104: R63–R67. SARS-CoV spike protein epitopes in unexposed individuals. Int. Immunol. 21:
4. Cameron, T. O., G. B. Cohen, S. A. Islam, and L. J. Stern. 2002. Examination of 63–71.
the highly diverse CD4(+) T-cell repertoire directed against an influenza peptide: 26. Kinnunen, T., A. Nieminen, W. W. Kwok, A. Närvänen, M. Rytkönen-Nissinen,
a step towards TCR proteomics. Immunogenetics 54: 611–620. S. Saarelainen, A. Taivainen, and T. Virtanen. 2010. Allergen-specific naı̈ve and
5. Newell, E. W., L. O. Klein, W. Yu, and M. M. Davis. 2009. Simultaneous de- memory CD4+ T cells exhibit functional and phenotypic differences between
tection of many T-cell specificities using combinatorial tetramer staining. Nat. individuals with or without allergy. Eur. J. Immunol. 40: 2460–2469.
Methods 6: 497–499. 27. Vingert, B., S. Perez-Patrigeon, P. Jeannin, O. Lambotte, F. Boufassa, F. Lemaı̂tre,
6. Song, Q., Q. Han, E. M. Bradshaw, S. C. Kent, K. Raddassi, B. Nilsson, W. W. Kwok, I. Theodorou, J. F. Delfraissy, J. Thèze, and L. A. ChakrabartiANRS
G. T. Nepom, D. A. Hafler, and J. C. Love. 2010. On-chip activation and sub- EP36 HIV Controllers Study Group. 2010. HIV controller CD4+ T cells respond to
sequent detection of individual antigen-specific T cells. Anal. Chem. 82: 473–477. minimal amounts of Gag antigen due to high TCR avidity. PLoS Pathog. 6: e1000780.
7. Day, C. L., N. P. Seth, M. Lucas, H. Appel, L. Gauthier, G. M. Lauer, 28. Cusick, M. F., M. Yang, J. C. Gill, and D. D. Eckels. 2011. Naturally occurring
G. K. Robbins, Z. M. Szczepiorkowski, D. R. Casson, R. T. Chung, et al. 2003. Ex CD4+ T-cell epitope variants act as altered peptide ligands leading to impaired
vivo analysis of human memory CD4 T cells specific for hepatitis C virus using helper T-cell responses in hepatitis C virus infection. Hum. Immunol. 72: 379–385.
MHC class II tetramers. J. Clin. Invest. 112: 831–842. 29. DeLong, J. H., K. H. Simpson, E. Wambre, E. A. James, D. Robinson, and
8. Scriba, T. J., M. Purbhoo, C. L. Day, N. Robinson, S. Fidler, J. Fox, J. N. Weber, W. W. Kwok. 2011. Ara h 1-reactive T cells in individuals with peanut allergy. J.
P. Klenerman, A. K. Sewell, and R. E. Phillips. 2005. Ultrasensitive detection and Allergy Clin. Immunol. 127: 1211–1218, e3.
phenotyping of CD4+ T cells with optimized HLA class II tetramer staining. J. 30. James, E. A., J. A. DeVoti, D. W. Rosenthal, L. J. Hatam, B. M. Steinberg,
Immunol. 175: 6334–6343. A. L. Abramson, W. W. Kwok, and V. R. Bonagura. 2011. Papillomavirus-specific
9. Chu, H. H., J. J. Moon, K. Takada, M. Pepper, J. A. Molitor, T. W. Schacker, CD4+ T cells exhibit reduced STAT-5 signaling and altered cytokine profiles in
K. A. Hogquist, S. C. Jameson, and M. K. Jenkins. 2009. Positive selection opti- patients with recurrent respiratory papillomatosis. J. Immunol. 186: 6633–6640.
mizes the number and function of MHCII-restricted CD4+ T cell clones in the 31. James, E. A., J. Bui, D. Berger, L. Huston, M. Roti, and W. W. Kwok. 2007.
naive polyclonal repertoire. Proc. Natl. Acad. Sci. USA 106: 11241–11245. Tetramer-guided epitope mapping reveals broad, individualized repertoires of tet-
10. Wambre, E., J. H. DeLong, E. A. James, R. E. Lafond, D. Robinson, and W. W. anus toxin-specific CD4+ T cells and suggests HLA-based differences in epitope
Kwok. 2012. Differentiation stage determines pathologic and protective allergen- recognition. Int. Immunol. 19: 1291–1301.
2482 BRIEF REVIEWS: MHC CLASS II TETRAMERS

32. Wang, S. F., L. Yao, S. J. Liu, P. Chong, W. T. Liu, Y. M. Chen, and J. C. Huang. 2010. Recurrence of type 1 diabetes after simultaneous pancreas-kidney trans-
2010. Identifying conserved DR1501-restricted CD4(+) T-cell epitopes in avian plantation, despite immunosuppression, is associated with autoantibodies and
H5N1 hemagglutinin proteins. Viral Immunol. 23: 585–593. pathogenic autoreactive CD4 T-cells. Diabetes 59: 947–957.
33. Yang, J., E. A. James, L. Huston, N. A. Danke, A. W. Liu, and W. W. Kwok. 2006. 52. Snir, O., M. Rieck, J. A. Gebe, B. B. Yue, C. A. Rawlings, G. Nepom,
Multiplex mapping of CD4 T cell epitopes using class II tetramers. Clin. Immunol. V. Malmström, and J. H. Buckner. 2011. Identification and functional character-
120: 21–32. ization of T cells reactive to citrullinated vimentin in HLA-DRB1*0401-positive
34. Novak, E. J., A. W. Liu, J. A. Gebe, B. A. Falk, G. T. Nepom, D. M. Koelle, and humanized mice and rheumatoid arthritis patients. Arthritis Rheum. 63: 2873–2883.
W. W. Kwok. 2001. Tetramer-guided epitope mapping: rapid identification and 53. Ráki, M., L. E. Fallang, M. Brottveit, E. Bergseng, H. Quarsten, K. E. Lundin, and
characterization of immunodominant CD4+ T cell epitopes from complex antigens. L. M. Sollid. 2007. Tetramer visualization of gut-homing gluten-specific T cells in
J. Immunol. 166: 6665–6670. the peripheral blood of celiac disease patients. Proc. Natl. Acad. Sci. USA 104: 2831–
35. Kwok, W. W., J. A. Gebe, A. Liu, S. Agar, N. Ptacek, J. Hammer, D. M. Koelle, 2836.
and G. T. Nepom. 2001. Rapid epitope identification from complex class-II- 54. Crawford, F., B. Stadinski, N. Jin, A. Michels, M. Nakayama, P. Pratt, P. Marrack,
restricted T-cell antigens. Trends Immunol. 22: 583–588. G. Eisenbarth, and J. W. Kappler. 2011. Specificity and detection of insulin-reactive
36. Worm, M., H. H. Lee, J. Kleine-Tebbe, R. P. Hafner, P. Laidler, D. Healey, CD4+ T cells in type 1 diabetes in the nonobese diabetic (NOD) mouse. Proc. Natl.
C. Buhot, A. Verhoef, B. Maillere, A. B. Kay, and M. Larche. 2011. Development Acad. Sci. USA 108: 16729–16734.
and preliminary clinical evaluation of a peptide immunotherapy vaccine for cat 55. Landais, E., P. A. Romagnoli, A. L. Corper, J. Shires, J. D. Altman, I. A. Wilson,
allergy. J. Allergy Clin. Immunol. 127: 89–97, 97.e1–14. K. C. Garcia, and L. Teyton. 2009. New design of MHC class II tetramers to
37. Danke, N. A., D. M. Koelle, C. Yee, S. Beheray, and W. W. Kwok. 2004. Auto- accommodate fundamental principles of antigen presentation. J. Immunol. 183:
reactive T cells in healthy individuals. J. Immunol. 172: 5967–5972. 7949–7957.
38. Danke, N. A., J. Yang, C. Greenbaum, and W. W. Kwok. 2005. Comparative study 56. Wong, R., R. Lau, J. Chang, T. Kuus-Reichel, V. Brichard, C. Bruck, and J. Weber.
of GAD65-specific CD4+ T cells in healthy and type 1 diabetic subjects. J. Auto- 2004. Immune responses to a class II helper peptide epitope in patients with stage
immun. 25: 303–311. III/IV resected melanoma. Clin. Cancer Res. 10: 5004–5013.
39. Mallone, R., S. A. Kochik, H. Reijonen, B. D. Carson, S. F. Ziegler, W. W. Kwok, 57. Aloysius, M. M., A. J. Mc Kechnie, R. A. Robins, C. Verma, J. M. Eremin,
and G. T. Nepom. 2005. Functional avidity directs T-cell fate in autoreactive CD4+ F. Farzaneh, N. A. Habib, J. Bhalla, N. R. Hardwick, S. Satthaporn, et al. 2009.
T cells. Blood 106: 2798–2805. Generation in vivo of peptide-specific cytotoxic T cells and presence of regulatory
40. Oling, V., J. Marttila, J. Ilonen, W. W. Kwok, G. Nepom, M. Knip, O. Simell, and T cells during vaccination with hTERT (class I and II) peptide-pulsed DCs. J.
H. Reijonen. 2005. GAD65- and proinsulin-specific CD4+ T-cells detected by Transl. Med. 7: 18.
MHC class II tetramers in peripheral blood of type 1 diabetes patients and at-risk 58. Slingluff, C. L., Jr., G. R. Petroni, W. C. Olson, M. E. Smolkin, M. I. Ross,
subjects. J. Autoimmun. 25: 235–243. N. B. Haas, W. W. Grosh, M. E. Boisvert, J. M. Kirkwood, and K. A. Chianese-

Downloaded from www.jimmunol.org on March 23, 2012


41. Veldman, C., R. Eming, S. Wolff-Franke, G. Sonderstrup, W. W. Kwok, and Bullock. 2009. Effect of granulocyte/macrophage colony-stimulating factor on cir-
M. Hertl. 2007. Detection of low avidity desmoglein 3-reactive T cells in pem- culating CD8+ and CD4+ T-cell responses to a multipeptide melanoma vaccine:
phigus vulgaris using HLA-DR beta 1*0402 tetramers. Clin. Immunol. 122: 330– outcome of a multicenter randomized trial. Clin. Cancer Res. 15: 7036–7044.
337. 59. Ayyoub, M., D. Dojcinovic, P. Pignon, I. Raimbaud, J. Schmidt, I. Luescher, and
42. Yang, J., N. Danke, M. Roti, L. Huston, C. Greenbaum, C. Pihoker, E. James, and D. Valmori. 2010. Monitoring of NY-ESO-1 specific CD4+ T cells using molec-
W. W. Kwok. 2008. CD4+ T cells from type 1 diabetic and healthy subjects exhibit ularly defined MHC class II/His-tag-peptide tetramers. Proc. Natl. Acad. Sci. USA
different thresholds of activation to a naturally processed proinsulin epitope. J. 107: 7437–7442.
Autoimmun. 31: 30–41. 60. Maus, M. V., J. L. Riley, W. W. Kwok, G. T. Nepom, and C. H. June. 2003. HLA
43. Long, S. A., M. R. Walker, M. Rieck, E. James, W. W. Kwok, S. Sanda, C. Pihoker, tetramer-based artificial antigen-presenting cells for stimulation of CD4+ T cells.
C. Greenbaum, G. T. Nepom, and J. H. Buckner. 2009. Functional islet-specific Clin. Immunol. 106: 16–22.
Treg can be generated from CD4+CD252 T cells of healthy and type 1 diabetic 61. Cochran, J. R., T. O. Cameron, and L. J. Stern. 2000. The relationship of MHC-
subjects. Eur. J. Immunol. 39: 612–620. peptide binding and T cell activation probed using chemically defined MHC class II
44. Durinovic-Belló, I., R. P. Wu, V. H. Gersuk, S. Sanda, H. G. Shilling, and oligomers. Immunity 12: 241–250.
G. T. Nepom. 2010. Insulin gene VNTR genotype associates with frequency and 62. Vollers, S. S., and L. J. Stern. 2008. Class II major histocompatibility complex
phenotype of the autoimmune response to proinsulin. Genes Immun. 11: 188–193. tetramer staining: progress, problems, and prospects. Immunology 123: 305–313.
45. James, E. A., A. K. Moustakas, J. Bui, G. K. Papadopoulos, G. Bondinas, 63. James, E. A., S. D. van Haren, R. A. Ettinger, K. Fijnvandraat, J. A. Liberman,
J. H. Buckner, and W. W. Kwok. 2010. HLA-DR1001 presents “altered-self” W. W. Kwok, J. Voorberg, and K. P. Pratt. 2011. T-cell responses in two unrelated
peptides derived from joint-associated proteins by accepting citrulline in three of its hemophilia A inhibitor subjects include an epitope at the factor VIII R593C mis-
binding pockets. Arthritis Rheum. 62: 2909–2918. sense site. J. Thromb. Haemost. 9: 689–699.
46. Mattapallil, M. J., P. B. Silver, J. J. Mattapallil, R. Horai, Z. Karabekian, 64. Fourneau, J. M., H. Cohen, and P. M. van Endert. 2004. A chaperone-assisted high
J. H. McDowell, C. C. Chan, E. A. James, W. W. Kwok, H. N. Sen, et al. 2011. yield system for the production of HLA-DR4 tetramers in insect cells. J. Immunol.
Uveitis-associated epitopes of retinal antigens are pathogenic in the humanized Methods 285: 253–264.
mouse model of uveitis and identify autoaggressive T cells. J. Immunol. 187: 1977– 65. Yang, J., A. Jaramillo, R. Shi, W. W. Kwok, and T. Mohanakumar. 2004. In vivo
1985. biotinylation of the major histocompatibility complex (MHC) class II/peptide
47. Raddassi, K., S. C. Kent, J. Yang, K. Bourcier, E. M. Bradshaw, V. Seyfert- complex by coexpression of BirA enzyme for the generation of MHC class II/tet-
Margolis, G. T. Nepom, W. W. Kwok, and D. A. Hafler. 2011. Increased fre- ramers. Hum. Immunol. 65: 692–699.
quencies of myelin oligodendrocyte glycoprotein/MHC class II-binding CD4 cells 66. Cunliffe, S. L., J. R. Wyer, J. K. Sutton, M. Lucas, G. Harcourt, P. Klenerman,
in patients with multiple sclerosis. J. Immunol. 187: 1039–1046. A. J. McMichael, and A. D. Kelleher. 2002. Optimization of peptide linker length
48. Pugliese, A., M. Zeller, A. Fernandez, Jr., L. J. Zalcberg, R. J. Bartlett, C. Ricordi, in production of MHC class II/peptide tetrameric complexes increases yield and
M. Pietropaolo, G. S. Eisenbarth, S. T. Bennett, and D. D. Patel. 1997. The insulin stability, and allows identification of antigen-specific CD4+T cells in peripheral
gene is transcribed in the human thymus and transcription levels correlated with blood mononuclear cells. Eur. J. Immunol. 32: 3366–3375.
allelic variation at the INS VNTR-IDDM2 susceptibility locus for type 1 diabetes. 67. Ge, X., J. A. Gebe, P. L. Bollyky, E. A. James, J. Yang, L. J. Stern, and
Nat. Genet. 15: 293–297. W. W. Kwok. 2010. Peptide-MHC cellular microarray with innovative data analysis
49. Chentoufi, A. A., and C. Polychronakos. 2002. Insulin expression levels in the system for simultaneously detecting multiple CD4 T-cell responses. PLoS ONE 5:
thymus modulate insulin-specific autoreactive T-cell tolerance: the mechanism by e11355.
which the IDDM2 locus may predispose to diabetes. Diabetes 51: 1383–1390. 68. Soen, Y., D. S. Chen, D. L. Kraft, M. M. Davis, and P. O. Brown. 2003. Detection
50. Laughlin, E., G. Burke, A. Pugliese, B. Falk, and G. Nepom. 2008. Recurrence of and characterization of cellular immune responses using peptide-MHC microarrays.
autoreactive antigen-specific CD4+ T cells in autoimmune diabetes after pancreas PLoS Biol. 1: E65.
transplantation. Clin. Immunol. 128: 23–30. 69. Mallone, R., S. A. Kochik, E. M. Laughlin, V. H. Gersuk, H. Reijonen,
51. Vendrame, F., A. Pileggi, E. Laughlin, G. Allende, A. Martin-Pagola, W. W. Kwok, and G. T. Nepom. 2004. Differential recognition and activation
R. D. Molano, S. Diamantopoulos, N. Standifer, K. Geubtner, B. A. Falk, et al. thresholds in human autoreactive GAD-specific T-cells. Diabetes 53: 971–977.

You might also like