Skip to main content

PERSPECTIVE article

Front. Immunol., 02 September 2015
Sec. Vaccines and Molecular Therapeutics

MHC Class II-Restricted Epitopes Containing an Oxidoreductase Activity Prompt CD4+ T Cells with Apoptosis-Inducing Properties

  • 1Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
  • 2ImCyse SA, Leuven, Belgium

Abrogating an unwanted immune response toward a specific antigen without compromising the entire immune system is a hoped-for goal in immunotherapy. Instead of manipulating dendritic cells and suppressive regulatory T cells, depleting effector T cells or blocking their co-stimulatory pathways, we describe a method to specifically inhibit the presentation of an antigen eliciting an unwanted immune reaction. Inclusion of an oxidoreductase motif within the flanking residues of MHC class II epitopes polarizes CD4+ T cells to cytolytic cells capable of inducing apoptosis in antigen presenting cells (APCs) displaying cognate peptides through MHC class II molecules. This novel function results from an increased synapse formation between both cells. Moreover, these cells eliminate by apoptosis bystander CD4+ T cells activated at the surface of the APC. We hypothesize that they would thereby block the recruitment of cells of alternative specificity for the same autoantigen or cells specific for another antigen associated with the pathology, providing a system by which response against multiple antigens linked with the same disease can be suppressed. These findings open the way toward a novel form of antigen-specific immunosuppression.

Introduction: A Distinct Subset of CD4+ T Cells

CD4+ T cells with cytotoxic activity have been occasionally described over the past 30 years (13). Their role in infection has been recognized, both during natural disease (46) and as an outcome of immunization (7, 8). Moreover, their antitumor potential seems to have been underestimated (9). Despite recent advances characterizing these cells as end-stage differentiated cells (10) and describing their cytolytic function through Fas–Fas ligand interaction and perforin/granzymes activity (11), the conditions under which they can be elicited remain poorly understood and their physiological relevance has not been fully established (12, 13).

CD4+ T cells can also differentiate into regulatory T cells (Tregs). Natural Tregs are selected in the thymus and show specificity for auto as well as alloantigens (14). The identification of Foxp3 as a master transcription regulator and subsequent gene profiling (15) have established their identity, with a phenotype, including constitutive expression of CD25, GITR, ICOS, CTLA-4, and CD103, production of IL-10, and absence of CD127 (16). Natural Tregs are essential for the control of autoimmunity and have shown a therapeutic potential in experimental autoimmune diseases and allograft rejection (17), but their use is still limited by difficulties of expanding stable populations, their lack of antigen specificity and concern of non-specific effect related to the production of suppressive cytokines (18).

Over recent years, a second category of Tregs has been described, called adaptive or induced Tregs (iTregs). These cells can be obtained both in vitro in absence of antigen by incubation with IL-10, vitamin D3, dexamethasone, or IFN-α (Tr1) and in vivo by antigen administration via the oral route (Th3) or by peptide administration in the absence of adjuvant via the transmucosal route (19) or subcutaneously (20). The molecular mechanisms underlying the function of iTregs are poorly understood, reflecting their heterogeneity. They do not express Foxp3, except for transient expression in the presence of TGF-β (21). Some iTregs overexpress T-bet when elicited by peptide administration without adjuvant (19), or GATA-3 after intranasal administration of ovalbumin (22), suggesting that the phenotype of iTregs and their functional properties vary according to initial T cell lineage commitment and degree of maturation (23). iTregs share an anergic status and limited capacities for in vitro expansion (18).

We describe here what we consider as a bona fide new and distinct functional subset of CD4+ T cells. The uniqueness of these cells are based on two key observations: one, these cells can be generated from any of the major subsets tested so far, including highly polarized Th1, Th2, and Th17 cells; two, in parallel to acquisition of apoptosis-inducing properties, such cells acquire a phenotype of terminally differentiated effector memory cells.

Induction of Cytolytic CD4+ T Cells

Formation of a synapse in between an antigen presenting cell (APC) expressing a MHC class II molecule presenting an antigen-derived epitope and a CD4+ T cell constitutes the earliest step of recognition by the adaptive immune system, and as such represents an ideal target for intervention.

The crystal structure of a MHC class II molecule shows that the cleft in which an epitope can be accommodated remains open on both sides, providing the opportunity to bind epitopes exceeding the sequence bound to the cleft. Thus, epitopes up to 20 amino acids have been described as being presented by class II molecules, although the sequence recognized by the TCR is limited to 8–9 amino acids. Retrospectively, it is surprising to realize that this general observation has attracted very limited attention. Residues located outside of the cleft, the so-called flanking residues, have been considered almost exclusively to identify their role in strengthening either the binding of the epitope in the cleft or alternatively the strength of TCR binding. Indeed, the nature of the amino acids just adjacent to positions P1 and P9, which correspond to the first and the last anchoring residues for the epitope, has been shown to influence such binding (24). This is all the more surprising as the strength of the synapse in between an APC and a CD4+ T cells is known to be one of the factors deciding upon the fate of the CD4+ T cells (25). Recent experiments have, however, demonstrated that modifications in flanking residues of a variety of epitopes can affect TCR recognition and CD4+ T cell function (26, 27).

Earlier research on one of the major class II-restricted T cell epitopes (p21–35) of the Der p 2 allergen derived from the house dust mite, Dermatophagoides pteronyssinus, showed that this peptide elicited CD4+ T cells, which appeared to eliminate APCs in culture (28, 29). The determinants in this peptide sequence that lead to this remarkable property were unknown. Extensive investigation of this epitope unveiled the presence of an intact amino terminal oxidoreductase motif (CxxS motif, where C stands for cysteine, S for serine, and x for any other amino acid) located within the flanking residues. This motif is characteristic of monocysteinic glutaredoxins (30), which are known to exert a nucleophilic attack on disulfide bridges and to create as such stable intermediates (31).

Amino acid substitution assays demonstrated that the presence of this motif in the Der p 2 (p21–35) sequence is essential for acquisition of cytolytic activities toward APCs (32). Introducing such motif in the flanking residues of other T cell epitopes demonstrates that the cytolytic properties are not limited to the specific Der p 2 epitope sequence context. The natural CxxS motif was further optimized by introducing a second cysteine, making a CxxC motif (33) presenting a higher oxidoreductase and reinforcing the cytolytic properties. This novel function results from an increased synapse formation between the APC and the antigen-specific CD4+ T cells. Carlier et al. further demonstrated that the target for the reduction was a constrained disulfide bridge located in the second extracellular domain of the CD4 molecule itself.

Strength of Synapse Formation and Acquisition of a Cytolytic Phenotype

The strength of the synapse is a determining factor for the fate of CD4+ T cells, low strength leads to ignorance or induction of anergy, while excessive strength could prime activation-induced cell death. The CD4 molecule binds via its first extracellular domain to residues located in the MHC class II molecule. By doing so, the synapse itself, which is by nature of low affinity, is physiologically strengthened. Reducing the disulfide bridge allows the formation of homodimers or polymers of CD4, thereby further strengthening the synapse, as demonstrated by increased number of doublets in between APCs and CD4+ T cells (32).

CD4 contains an intracellular domain with subdomains for the recruitment of kinases associated with early signaling. Exposure of naïve CD4+ T cells to epitopes with a CxxC motif located in flanking residues results in increased rates of recruitment of Lck and subsequently ZAP70, forming an activation complex with CD3. An increase in the kinetics and extent of CD3ζ phosphorylation and degradation further indicated that the increase in the activation followed a physiological pathway. Furthermore, we observed that the increased rate of this early signaling complex was followed by sustained activation of PI3K and AKT. Phosphorylation of AKT is a crucial event for cells, as it drives the proliferation capacity of the cells, their metabolism, and phenotype adoption. Thus, increased phosphorylation of AKT resulted in prevention of nuclear migration of FOXO-3a, triggered the aerobic glycolytic pathway shared by effector cells and activated the mTOR complex pathway (our unpublished data). Of much interest, these activation and metabolic properties are opposite to the ones observed with Tregs, in which AKT activation is reduced and the metabolism switched to oxidated lipid phosphorylation, indicating already that on phenotypical grounds, cells exposed to epitopes containing the CxxC motif adopted a diametrically opposite phenotype as Tregs do.

Functionally speaking, cells exposed to epitopes containing a CxxC motif promptly adopted a phenotype of effector memory cells, characterized by rapid loss of expression of CD62L and high expression of CD44. Other relevant surface markers include CD25, a characteristic shared with Tregs, and which provides cells with the possibility of utilizing IL-2 when present at low concentrations leading to a proliferative advantage over alternative effector cells. Low expression of CD28 and increased expression of CTLA-4 are also shared features with Tregs. At the transcriptional level, cells are characterized by co-expression of T-bet and GATA-3, considered as being exclusive from each other in effector cells, Th1 expressing T-bet and Th2 cells expressing GATA-3. Worth mentioning is that, under no circumstances, it was possible to detect Foxp3, at protein or even mRNA level (Malek Abrahimians et al., in preparation). The production of cytokines is essentially limited to IFN-γ. FasL and granzyme B are transcribed and the proteins produced. Both actively participate in the induction of apoptosis of target cells, such as APCs with which a synapse is formed. Perforin can be transcribed but seemingly with no production of the protein. Blocking experiments have confirmed the direct participation of both granzyme B and FasL in the cytolytic activity of the cells, while inhibitors of perforin showed no effect (32).

Conversion of Polarized Cells and Induction of Bystander CD4+ T Cell Apoptosis

We observed that polarized cells exposed to their cognate epitope in the presence of a CxxC motif progressively lose their phenotypic characteristics to acquire cytolytic properties. Interestingly enough, once the cytolytic properties are acquired, cells seemingly do not revert to their initial phenotype even under highly polarizing conditions (Carlier et al., in preparation).

The question was extended to bystander CD4+ T cells to determine whether the technology would be effective in a multi-epitope and multi-antigen pathology; in other words, whether it was possible to suppress a polyclonal CD4+ T cell response using a single epitope from a single antigen. We demonstrated that CD4+ T cells that had acquired cytolytic potential, were capable of eliminating by apoptosis whatever CD4+ T cells, provided these cells were activated at the surface of the same APC. This apoptosis occurred independently of the elimination of the APC itself (32).

Taken altogether, the combination of the possibility to convert naïve or polarized cells into a cytolytic phenotype, with the possibility to eliminate by apoptosis activated bystander CD4+ T cells, constitutes a comprehensive approach with potential for therapeutic intervention.

Pre-Clinical Evidence toward a Therapeutic Application

In a model of experimental allergic asthma, as induced by nasal instillation of recombinant allergens prepared from the house dust mite, D. pteronyssinus, immunization with a peptide encompassing a class II-restricted epitope of either Der p 1 or Der p 2 was sufficient to eliminate the bronchial reactivity to exposure not only to the allergen serving to induce asthma but also to an alternative allergen. Besides, and more importantly, such a treatment eliminated the bronchial reactivity to a non-specific stimulus largely used in a clinical setting, namely acetylcholine, demonstrating that such an approach is able to reduce both specific and non-specific bronchial reactivity.

In a model of skin graft rejection, pre-immunization of female C57BL/6 mice with a single epitope derived from a single antigen encoded by the Y chromosome was sufficient to obtain full tolerance to the grafting of a male skin. This model also established that mice having rejected a graft, and therefore under condition of higher reactivity to alloantigens were rendered tolerant to a subsequent graft if pretreated with the epitope of the Dby antigen. Additionally, mice pre-immunized and having tolerated a first graft were tolerant to a second identical graft 4 months later and with no intermediate treatment, providing an indication of the in vivo persistence of such Dby-specific cytolytic CD4+ T cells.

In a model of multiple sclerosis, either pre-immunization with a class II-restricted epitope from the MOG protein (a constituent of the myelin gain) or immunization initiated weeks after disease induction, it was possible to prevent the appearance of clinical signs or, to some extent, reduce such signs. Strikingly, in such a model, gains of myelin were reconstituted in a context in which it could be observed that the inflammatory infiltrates in the central nervous system white matter or the spinal cord had been completely eliminated. Interestingly, infusion of cells, in the same model, converted in vitro to a cytolytic phenotype was efficient to either prevent or suppress the disease. Noticeably, a clinical trial with patients with relapsing–remitting multiple sclerosis has been recently initiated using the same principle.

In a model of gene therapy, in which the immune response toward the viral vector prohibits an effective administration and expression of a transgene, we have demonstrated that administration of a peptide encompassing a class II-restricted epitope of hexon 6, a capsid protein of adenovirus 5, was sufficient to prevent such an immune response, allowing re-administration of the same construct, use of a significantly lower dose of construct, use of improved transgenes, and prolongation of transgene expression. Considering that, in terms of immunogenicity, adenovirus represents the worst-case scenario, we surmise that alternative viruses, such as the adeno-associated virus (AAV), would be amenable to the same approach. Importantly, the same method could be used for gene vaccination.

In the spontaneous non-obese diabetes (NOD) mouse model of type 1 diabetes, immunization with a MHC class II-restricted epitope derived from glutamic acid decarboxylase (GAD65), a major autoantigen, prevents autoimmune destruction of pancreatic β-cells. Furthermore, passive transfer of in vitro derived cytolytic CD4+ T cells also prevented diabetes. Interestingly, this setting confirms that immunization with a single epitope is sufficient to prevent autoimmunity toward multiple antigens implicated in the disease process.

Risk Assessment

There is a theoretical risk of reversibility or conversion of cytolytic CD4+ T cells to pathogenic effector cells. However, efforts to force this under stringent in vitro conditions have been unsuccessful (our unpublished data). Furthermore, the number of generated cytolytic cells is very limited as compared to bulk of pathogenic effector cells present in the setting of an ongoing disease, indicating that, even in case of reversibility toward an effector phenotype, the clinical relevance of this would not be perceptible.

Although the concept makes use of a single epitope selected from an antigen directly implicated in the disease under investigation, the very fact that cytolytic CD4+ T cells eliminate activated bystander CD4+ T cells could, in theory, lead to elimination of useful effector cells toward, for instance, a microorganism. However, as the activation and cytolytic properties of the cells requires the formation of a synapse with a cognate epitope, we have to envision a situation in which all the immune response toward the microorganism is elaborated at the same time and in the same location as the response that has to be eliminated. In a large majority of the case, and in particular in organ-specific autoimmune diseases, the pathogenic response is elaborated in the draining regional lymph nodes, which is not likely to be the port of entry of the microorganism. We therefore deem that the likelihood of increasing the susceptibility to a concomitant infection is remote.

Any intervention modifying the fate of a cell carries the risk of increasing tumorigenic potential. However, to evaluate this risk, we have analyzed the caryotype of these transformed cells and have not identified any significant alterations in comparison to effector cells under recurrent stimulation. Moreover, measurement of the telomerase activity to evaluate the status of senescence of such cells has not showed reduced activity (our unpublished data).

Oxidoreductase-Containing Versus Unmodified Epitopes

Peptides encompassing class II-restricted epitopes have been used in several settings in an attempt to control disease processes. Such peptides are administered via different routes, often transmucosal, and in absence of adjuvant. Some results have been observed in animal models, yet very little if any in the clinic (34).

The mechanism of action of such unmodified epitopes is related to restimulation-induced cell death (RICD) (35). This mechanism is linked to the extrinsic pathway of apoptosis induction, a mechanism associated with signaling through the TNFR superfamily, including Fas–FasL interaction and the formation of DISC, the death-inducing signaling complex, recruiting and activating caspases 8 and 10. This is opposed to the intrinsic pathway in which depolarization of the mitochondrial membrane activates caspase 9. RICD prevents uncontrolled expansion of an immune response and is responsible for the contraction phase following an immune response, in which the number of activated CD4+ T cells drop dramatically, to maintain a small population entering into a cycle of memory phenotype conversion.

The results obtained in animal experiments using unmodified epitopes administered without adjuvant is deemed to depend on the elimination, by the induction of apoptosis by the extrinsic pathway, of the CD4+ T lymphocytes specific only for the very antigen from which the epitope has been selected. This explains the significant, though partial beneficial effect of such approach. However, in a clinical setting, wherein a large number of antigens are involved, the elimination of a single CD4+ T cell population will provide very limited, if any benefit, very much in keeping with the absence of clinical efficacy (36).

In the present setting, instead of reducing the size of a single CD4+ T cell population specific for an epitope of a single antigen, the intrinsic pathway of apoptosis induction is used in combination with the extrinsic pathway. Effector cells utilizing both the intrinsic and extrinsic apoptosis induction pathways provide an active method to eliminate effector CD4+ T cells of alternative specificity (alternative epitopes of the same antigen and alternative antigens) (37), i.e., eliminating a much larger spectrum of effector cells associated with a disease process than unmodified epitopes. Besides, elimination of the APCs with which the synapse is formed provides a means to prevent the recruitment of new cells in the pathological process. These fundamental differences in between the mode of action of cytolytic CD4+ T cells and the mere depletion of single effector cell population establish the superiority of the approach.

Concluding Remarks

These pre-clinical observations on oxidoreductase-containing epitopes and their elicitation of CD4+ T cells capable of specifically eliminating APCs presenting the nominal peptide opens a door to a novel therapeutic strategy in immune disease and leads to hypothesize that any class II-restricted T cell epitope can be modified by addition of a oxidoreductase motif in order to induce specific CD4+ T cells with apoptosis-inducing properties and thus overcome any unwanted class II-restricted immune response. The high homology in between the synapse created at MHC class II level in mouse and in human leads to the assumption that the observations carried out in pre-clinical models could be extrapolated to human cells. Indeed, we have demonstrated efficacy in converting in vitro human CD4+ T cells to a cytolytic phenotype, allowing application to human disease in the near future.

Conflict of Interest Statement

This research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Feighery C, Stastny P. HLA-D region-associated determinants serve as targets for human cell-mediated lysis. J Exp Med (1979) 149(2):485–94. doi:10.1186/1423-0127-20-60

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Fleischer B. Acquisition of specific cytotoxic activity by human T4+ T lymphocytes in culture. Nature (1984) 308(5957):365–7. doi:10.1038/308365a0

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Gromkowski SH, Hepler KM, Janeway CA Jr. Low doses of interleukin 2 induce bystander cell lysis by antigen-specific CD4+ inflammatory T cell clones in short-term assay. Eur J Immunol (1988) 18(9):1385–9. doi:10.1002/eji.1830180913

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Adhikary D, Behrends U, Moosmann A, Witter K, Bornkamm GW, Mautner J. Control of Epstein-Barr virus infection in vitro by T helper cells specific for virion glycoproteins. J Exp Med (2006) 203(4):995–1006. doi:10.1084/jem.20051287

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Swain SL, Agrewala JN, Brown DM, Jelley-Gibbs DM, Golech S, Huston G, et al. CD4+ T-cell memory: generation and multi-faceted roles for CD4+ T cells in protective immunity to influenza. Immunol Rev (2006) 211:8–22. doi:10.1038/nm.2142

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Brown DM. Cytolytic CD4 cells: direct mediators in infectious disease and malignancy. Cell Immunol (2010) 262(2):89–95. doi:10.1016/j.cellimm.2010.02.008

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Woodland DL, Hogan RJ, Zhong W. Cellular immunity and memory to respiratory virus infections. Immunol Res (2001) 24(1):53–67. doi:10.1385/IR:24:1:53

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Tsuji M, Romero P, Nussenzweig RS, Zavala F. CD4+ cytolytic T cell clone confers protection against murine malaria. J Exp Med (1990) 172(5):1353–7. doi:10.1084/jem.172.5.1353

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Perez-Diez A, Joncker NT, Choi K, Chan WF, Anderson CC, Lantz O, et al. CD4 cells can be more efficient at tumor rejection than CD8 cells. Blood (2007) 109(12):5346–54. doi:10.1182/blood-2006-10-051318

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Appay V, Zaunders JJ, Papagno L, Sutton J, Jaramillo A, Waters A, et al. Characterization of CD4(+) CTLs ex vivo. J Immunol (2002) 168(11):5954–8. doi:10.4049/jimmunol.168.11.5954

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Hahn S, Gehri R, Erb P. Mechanism and biological significance of CD4-mediated cytotoxicity. Immunol Rev (1995) 146:57–79. doi:10.1111/j.1600-065X.1995.tb00684.x

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Cheroutre H, Husain MM. CD4 CTL: living up to the challenge. Semin Immunol (2013) 25(4):273–81. doi:10.1016/j.smim.2013.10.022

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Soghoian DZ, Streeck H. Cytolytic CD4(+) T cells in viral immunity. Expert Rev Vaccines (2010) 9(12):1453–63. doi:10.1586/erv.10.132

PubMed Abstract | CrossRef Full Text | Google Scholar

14. von Herrath MG, Harrison LC. Antigen-induced regulatory T cells in autoimmunity. Nat Rev Immunol (2003) 3(3):223–32. doi:10.1038/nri1029

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Knoechel B, Lohr J, Zhu S, Wong L, Hu D, Ausubel L, et al. Functional and molecular comparison of anergic and regulatory T lymphocytes. J Immunol (2006) 176(11):6473–83. doi:10.4049/jimmunol.176.11.6473

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Liu W, Putnam AL, Xu-Yu Z, Szot GL, Lee MR, Zhu S, et al. CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med (2006) 203(7):1701–11. doi:10.1084/jem.20060772

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Sakaguchi S, Sakaguchi N, Shimizu J, Yamazaki S, Sakihama T, Itoh M, et al. Immunologic tolerance maintained by CD25+ CD4+ regulatory T cells: their common role in controlling autoimmunity, tumor immunity, and transplantation tolerance. Immunol Rev (2001) 182:18–32. doi:10.1034/j.1600-065X.2001.1820102.x

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Bluestone JA. Regulatory T-cell therapy: is it ready for the clinic? Nat Rev Immunol (2005) 5(4):343–9. doi:10.1038/nri1574

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Anderson PO, Manzo BA, Sundstedt A, Minaee S, Symonds A, Khalid S, et al. Persistent antigenic stimulation alters the transcription program in T cells, resulting in antigen-specific tolerance. Eur J Immunol (2006) 36(6):1374–85. doi:10.1002/eji.200635883

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Verhoef A, Alexander C, Kay AB, Larché M. T cell epitope immunotherapy induces a CD4+ T cell population with regulatory activity. PLoS Med (2005) 2(3):e78. doi:10.1371/journal.pmed.0020078

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity (2006) 24(2):179–89. doi:10.1016/j.immuni.2006.01.001

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Akbari O, Freeman GJ, Meyer EH, Greenfield EA, Chang TT, Sharpe AH, et al. Antigen-specific regulatory T cells develop via the ICOS-ICOS-ligand pathway and inhibit allergen-induced airway hyperreactivity. Nat Med (2002) 8(9):1024–32. doi:10.1038/nm745

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Cobbold SP. The hidden truth about gene expression in Tregs: is it what you don’t see that counts? Eur J Immunol (2006) 36(6):1360–3. doi:10.1002/eji.200636171

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Lovitch SB, Pu Z, Unanue ER. Amino-terminal flanking residues determine the conformation of a peptide-class II MHC complex. J Immunol (2006) 176(5):2958–68. doi:10.4049/jimmunol.176.5.2958

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Purvis HA, Stoop JN, Mann J, Woods S, Kozijn AE, Hambleton S, et al. Low-strength T-cell activation promotes Th17 responses. Blood (2010) 116(23):4829–37. doi:10.1182/blood-2010-03-272153

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Carson RT, Vignali KM, Woodland DL, Vignali DA. T cell receptor recognition of MHC class II-bound peptide flanking residues enhances immunogenicity and results in altered TCR V region usage. Immunity (1997) 7(3):387–99. doi:10.1016/S1074-7613(00)80360-X

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Holland C, Cole D, Godkin A. Re-directing CD4+ T cell responses with the flanking residues of MHC class II-bound peptides: the core is not enough. Front Immunol (2013) 4:172. doi:10.3389/fimmu.2013.00172

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Wu B, Vander Elst L, Carlier V, Jacquemin M, Saint-Remy JM. Dermatophagoides pteronyssinus group 2 allergen contains a universally immunogenic T cell epitope. J Immunol (2002) 169:2430. doi:10.4049/jimmunol.169.5.2430

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Janssens W, Carlier V, Wu B, VanderElst L, Jacquemin M, Saint-Remy JM. CD4+CD25+ T cells lyse antigen-presenting B cells by Fas-Fas ligand interaction in an epitope-specific manner. J Immunol (2003) 171:4604–12. doi:10.4049/jimmunol.171.9.4604

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Meyer Y, Buchanan BB, Vignols F, Reichheld JP. Thioredoxins and glutaredoxins: unifying elements in redox biology. Annu Rev Genet (2009) 296:55–63. doi:10.1146/annurev-genet-102108-134201

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Schwertassek U, Balmer Y, Gutscher M, Weingarten L, Preuss M, Engelhard J, et al. Selective redox regulation of cytokine receptor signaling by extracellular thoredoxin-1. EMBO J (2007) 26(13):3086–97. doi:10.1038/sj.emboj.7601746

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Carlier V, Vander Elst L, Janssens W, Jacquemin MG, Saint-Remy JM. Increased synapse formation obtained by T cell epitopes containing a CxxC motif in flanking residues convert CD4+ T cells into cytolytic effectors. PLoS One (2012) 7(10):e45366. doi:10.1371/journal.pone.00453

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Schmidt B, Ho L, Hogg PJ. Allosteric disulfide bonds. Biochemistry (2006) 45(24):7429–33. doi:10.1021/bi0603064

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Anderson R, Jabri B. Vaccine against autoimmune disease: antigen-specific immunotherapy. Curr Opin Immunol (2013) 25(3):410–7. doi:10.1016/j.coi.2013.02.004

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Snow A, Pandiyan P, Zheng L, Krummey S, Lenardo M. The power and the promise of restimulation-induced cell death in human immune diseases. Immunol Rev (2010) 236:68–82. doi:10.1111/j.1600-065X.2010.00917.x

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Nicholas D, Odumosu O, Langridge WH. Autoantigen based vaccines for type 1 diabetes. Discov Med (2011) 11(59):293–301.

PubMed Abstract | Google Scholar

37. Prasad S, Kohm AP, McMahon JS, Luo X, Miller SD. Pathogenesis of NOD diabetes is initiated by reactivity to the insulin B chain 9-23 epitope and involves functional epitope spreading. J Autoimmun (2012) 39:347–53. doi:10.1016/j.jaut.2012.04.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: MHC class II epitopes, oxidoreductase activity, cytolytic CD4+ T cells, apoptosis, immune suppression

Citation: Malek Abrahimians E, Carlier VA, Vander Elst L and Saint-Remy J-MR (2015) MHC class II-restricted epitopes containing an oxidoreductase activity prompt CD4+ T cells with apoptosis-inducing properties. Front. Immunol. 6:449. doi: 10.3389/fimmu.2015.00449

Received: 12 June 2015; Accepted: 18 August 2015;
Published: 02 September 2015

Edited by:

Brian J. Czerniecki, University of Pennsylvania, USA

Reviewed by:

Urszula Krzych, Walter Reed Army Institute of Research, USA
Daniel Olive, INSERM UMR 891 Institut Paoli Calmettes, France

Copyright: © 2015 Malek Abrahimians, Carlier, Vander Elst and Saint-Remy. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jean-Marie R. Saint-Remy, ImCyse, Bioincubator II, Gaston Geenslaan 1, Leuven 3001, Belgium, jean-marie.saint-remy@med.kuleuven.be

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.